201
|
Budagyan K, Cannon AC, Chatoff A, Benton D, Kurimchak AM, Araiza-Olivera D, Gerasimova A, Snyder NW, Duncan JS, Uribe-Alvarez C, Chernoff J. KRAS G12V mutation-selective requirement for ACSS2 in colorectal adenoma formation. Cell Rep 2025; 44:115444. [PMID: 40131933 DOI: 10.1016/j.celrep.2025.115444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 02/03/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Oncogenic KRAS mutations are prevalent in colorectal cancer (CRC) and linked to poor prognosis and therapeutic resistance. Emerging evidence suggests that specific KRAS mutations differentially influence treatment responses. In this study, we generate isogenic Apc-null mouse colon epithelial cells with four common KRAS mutations. Transcriptomic and proteomic analyses reveal significant enrichment of cholesterol and lipid metabolism pathways in KRAS G12V cells, driven by increased SREBP1 expression and mTORC1 activation. Furthermore, KRAS G12V cells exhibit elevated ACSS2 expression and greater dependence on ACSS2 for proliferative advantage compared to other mutants. Inhibition of ACSS2 uniquely sensitizes KRAS G12V cells to MEK inhibition, highlighting a distinct therapeutic vulnerability. Finally, ACSS2 plays a critical role in early KRAS G12V adenoma development, unlike in KRAS G12D adenomas. These findings highlight mutation-specific metabolic reprogramming in KRAS-driven CRC and identify ACSS2 as a potential therapeutic target.
Collapse
Affiliation(s)
- Konstantin Budagyan
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Alexa C Cannon
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Adam Chatoff
- Department of Cancer & Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Dorothy Benton
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Alison M Kurimchak
- Cancer Signaling & Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Daniela Araiza-Olivera
- Cancer Signaling & Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Anastasiia Gerasimova
- Cancer Signaling & Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Nathaniel W Snyder
- Department of Cancer & Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Cancer Signaling & Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - James S Duncan
- Cancer Signaling & Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Cristina Uribe-Alvarez
- Cancer Signaling & Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| | - Jonathan Chernoff
- Cancer Signaling & Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
202
|
Yu R, Ji X, Zhang P, Zhang H, Qu H, Dong W. The potential of chimeric antigen receptor -T cell therapy for endocrine cancer. World J Surg Oncol 2025; 23:153. [PMID: 40264184 PMCID: PMC12012980 DOI: 10.1186/s12957-025-03745-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/07/2025] [Indexed: 04/24/2025] Open
Abstract
Endocrine cancer, a relatively rare and heterogeneous tumor with diverse clinical features. The facile synthesis of hormones further complicates endocrine cancer treatment. Thus, the development of safe and effective systemic treatment approaches, such as chimeric antigen receptor (CAR) T cell therapy, is imperative to enhance the prognosis of patients with endocrine cancer. Although this therapy has achieved good results in the treatment of hematological malignancies, it encounters diverse complications and challenges in the context of endocrine cancer. This review delineates the generation of CAR-T cells, examines the potential of CAR-T cell therapy for endocrine cancer, enumerates pivotal antigens linked to endocrine cancer, encapsulates the challenges confronted with CAR-T cell therapy for endocrine cancer, and expounds upon strategies to overcome these limitations. The primary objective is to provide insightful perspectives that can contribute to the advancement of CAR-T cell therapy in the field of endocrine cancer.
Collapse
Affiliation(s)
- Ruonan Yu
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Xiaoyu Ji
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Ping Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Hao Zhang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China
| | - Huiling Qu
- Department of Neurology, The General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, Liaoning, 110840, China.
| | - Wenwu Dong
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 Nanjing North Street, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
203
|
Zhao S, Sun D, Yu H, Wang M, Xu B, Wang Y, Hu F, Wang X, Zhang J, Wang Y, Chai J. Oxaliplatin accelerates immunogenic cell death by activating the cGAS/STING/TBK1/IRF5 pathway in gastric cancer. FEBS J 2025. [PMID: 40260556 DOI: 10.1111/febs.70102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 11/11/2024] [Accepted: 04/07/2025] [Indexed: 04/23/2025]
Abstract
Immunogenic cell death is a tumor cell death involving both innate and adaptive immune responses. Given the published findings that oxaliplatin causes the secretion of high mobility group box 1 (HMGB1) from cancer cells, which is necessary for the initiation of immunogenic cell death, we investigated whether oxaliplatin plays an anticancer role in gastric cancer by inducing immunogenic cell death and further explored its mechanism. We found that oxaliplatin inhibited viability and induced pyroptosis, immunogenic cell death, the production of reactive oxygen species, mitochondrial permeability transition pore (mPTP) opening, and cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) axis activation in gastric cancer cells. Suppressing mPTP opening (cyclosporine treatment), depleting mitochondrial DNA (mtDNA; ethidium bromide treatment), or STING downregulation (H151 or si-STING treatment) reversed cGAS/STING pathway activation and the increased immunogenic cell death induced by oxaliplatin in MKN-45 and AGS cells. Moreover, oxaliplatin induced immunogenic cell death via activating the cGAS/STING/TANK-binding kinase 1 (TBK1; also known as serine/threonine-protein kinase TBK1)/interferon regulatory factor 5 (IRF5) pathway. In conclusion, oxaliplatin treatment could induce immunogenic cell death and mPTP opening and activate the cGAS/STING/TBK1/IRF5 pathway in gastric cancer cells.
Collapse
Affiliation(s)
- Siwei Zhao
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Dong Sun
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Hang Yu
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Menglin Wang
- Department of Plastic Surgery, The First Affiliated Hospital, Dalian Medical University, China
| | - Botao Xu
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yufei Wang
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Fangqi Hu
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaofeng Wang
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jiazi Zhang
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yongsheng Wang
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Chai
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
204
|
Nguyen-Hoang N, Nazzal M, Schneider BP, Trivedi MS, Hertz DL. Review of the contribution of clinical and genetic factors to the racial disparity in taxane-induced peripheral neuropathy. Crit Rev Oncol Hematol 2025; 211:104739. [PMID: 40274246 DOI: 10.1016/j.critrevonc.2025.104739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/24/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025] Open
Abstract
Taxanes are first-line chemotherapy for several solid tumors, but their use is often limited by taxane-induced peripheral neuropathy (TIPN), which can cause acute symptoms in up to 70 % of patients and severely deteriorate long-term quality of life. Recent evidence from large prospective observational studies confirms a dramatic racial disparity, with Black/African-ancestry patients facing roughly two times greater risk of TIPN compared to White patients. Understanding the root causes of this disparity is a critical first step toward eliminating inequities in cancer treatment side effects, aligning with a major goal of the U.S. National Cancer Institute's National Cancer Plan. This review examines clinical and genetic factors contributing to racial differences in TIPN, focusing on those that have been associated with TIPN risk and are more prevalent within Black/African-ancestry individuals. Pre-existing neuropathy, vitamin D insufficiency, metabolic risk factors (obesity/diabetes), systemic taxane exposure, and genetic variants are discussed as potential contributors to this racial disparity. The review concludes by describing additional research that is needed to determine which of these factors are responsible for this disparity and what types of translational clinical studies could be conducted to target these mechanisms and reduce inequity. These findings could inform clinical strategies that improve long-term quality of life and promote health equity in taxane-treated cancer patients in the U.S. and globally.
Collapse
Affiliation(s)
- Nam Nguyen-Hoang
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA
| | - Maisa Nazzal
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA
| | - Bryan P Schneider
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
| | - Meghna S Trivedi
- Herbert Irving Comprehensive Cancer Center/Columbia University, New York, NY 10032, USA
| | - Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA; University of Michigan Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI 48109, USA.
| |
Collapse
|
205
|
Michaels L, Noor M, Aslam T. Clinical and imaging strategies for the assessment of the ocular side effects of systemic targeted anti-cancer therapies. Eur J Cancer 2025; 222:115452. [PMID: 40306116 DOI: 10.1016/j.ejca.2025.115452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
Systemic targeted anti-cancer therapies selectively target cancerous cells whilst limiting systemic side effects. The eye however, is a particularly sensitive organ and the expanding use of the newer targeted chemotherapy agents has been associated with multiple ocular side effects. In this review we provide an update of the ocular side effects of the newer targeted chemotherapy agents along with suggested minimum, pragmatic, evidence-based strategies for effective screening or monitoring for potential ocular side effects. This framework is designed to guide oncologists, trial managers, protocol developers and regulatory authorities so that appropriate ophthalmic clinical examinations and non-invasive modern imaging can be requested and commissioned according to a patient's specific treatment.
Collapse
Affiliation(s)
- Luke Michaels
- St Thomas' Hospital, Westminster Bridge Road, London SE1 7EH, United Kingdom
| | - Maha Noor
- Manchester Royal Eye Hospital, Oxford Road, Manchester M13 9WL, United Kingdom
| | - Tariq Aslam
- Manchester Royal Eye Hospital, Oxford Road, Manchester M13 9WL, United Kingdom; School of Health Sciences, University of Manchester, Oxford Road, Manchester M139PL, United Kingdom.
| |
Collapse
|
206
|
Zhao X, Li Y, Zhang H, Cai Y, Wang X, Liu Y, Li T, Xu C, Teng Y, Li D, Li F. PAK5 promotes the trastuzumab resistance by increasing HER2 nuclear accumulation in HER2-positive breast cancer. Cell Death Dis 2025; 16:323. [PMID: 40258843 PMCID: PMC12012021 DOI: 10.1038/s41419-025-07657-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/23/2025]
Abstract
Nuclear HER2 (N-HER2) predicts resistance to HER2-targeted therapy and poor prognosis of breast cancer patients, and the underlying mechanisms remain unclear. Here, we show that high expression of p21-activated kinase 5 (PAK5) is associated with HER2-targeted therapy resistance and poor outcomes of breast cancer patients. Excitingly, we find an increase in N-HER2 protein expression in patients with high PAK5 expression, who demonstrate resistance to trastuzumab treatment. PAK5 phosphorylates methyltransferase METTL14 on serine 399 to enhance m6A modification of lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), leading to increased MALAT1 stability. The stabilized MALAT1 inhibits ubiquitin-proteasomal degradation of the N-HER2 by affecting the interaction of deubiquitinase USP8 and N-HER2, thereby promoting N-HER2 accumulation. Moreover, HER2 upregulates the expression of PAK5 and MALAT1, activating the HER2-MALAT1 positive feedback loop. Importantly, PAK5 promotes the therapeutic resistance of HER2-positive breast cancer cells by increasing N-HER2 protein both in vitro and vivo. These findings highlight PAK5 as a therapeutic target for combating trastuzumab resistance in HER2-positive breast cancer.
Collapse
MESH Headings
- Humans
- p21-Activated Kinases/metabolism
- p21-Activated Kinases/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Trastuzumab/pharmacology
- Trastuzumab/therapeutic use
- Female
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- RNA, Long Noncoding/metabolism
- RNA, Long Noncoding/genetics
- Animals
- Cell Line, Tumor
- Mice
- Mice, Nude
- Gene Expression Regulation, Neoplastic/drug effects
- Cell Nucleus/metabolism
- Cell Nucleus/drug effects
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Xin Zhao
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China.
| | - Hongyan Zhang
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Yihang Cai
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Xu Wang
- Department of Breast Surgery, Department of Surgical Oncology, Research Unit of General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yidu Liu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Tingting Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Chendong Xu
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China
| | - Yuee Teng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.
| | - Danni Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.
| | - Feng Li
- Department of Cell Biology, Key Laboratory of Cell Biology, National Health Commission of the PRC and Key Laboratory of Medical Cell Biology, Ministry of Education of the PRC, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
207
|
Liu Z, Lenz HJ, Yu J, Zhang L. Differential Response and Resistance to KRAS-Targeted Therapy. Mol Carcinog 2025. [PMID: 40256920 DOI: 10.1002/mc.23908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/10/2025] [Indexed: 04/22/2025]
Abstract
KRAS is the most frequently mutated oncogene. In epithelial malignancies such as lung, colorectal, and pancreatic tumors, KRAS is mutated in 25 to above 90% cases. KRAS was considered undruggable for over three decades until the recent development of covalent inhibitors targeting the KRAS G12C mutant. The recent approval of the KRAS G12C inhibitors sotorasib and adagrasib has ushered in a new era of KRAS-targeted therapy. Despite this success, a major challenge in KRAS-targeted therapy is intrinsic and acquired resistance to KRAS inhibitors. Clinical studies have shown that many patients with KRAS G12C cancers did not respond to sotorasib and adagrasib. Colorectal cancer, in particular, has a markedly lower response rate to KRAS G12C inhibitors compared to non-small cell lung cancer. Furthermore, the therapeutic response to KRAS G12C inhibition was short-lived, with quick emergence of acquired resistance. In this review, we summarize several major themes that have emerged from recent clinical and preclinical studies on the mechanisms of intrinsic and acquired resistance to KRAS-targeted therapy in colorectal, lung, and pancreatic cancers. We also discuss various combination strategies for targeting these mechanisms to overcome resistance to KRAS inhibitors.
Collapse
Affiliation(s)
- Zhaojin Liu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Heinz-Josef Lenz
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Jian Yu
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Lin Zhang
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, California, USA
- Keck School of Medicine of USC, Norris Comprehensive Cancer Center, Los Angeles, California, USA
| |
Collapse
|
208
|
Davis E, Ermi AG, Sarkar D. Astrocyte Elevated Gene-1/Metadherin (AEG-1/MTDH): A Promising Molecular Marker and Therapeutic Target for Hepatocellular Carcinoma. Cancers (Basel) 2025; 17:1375. [PMID: 40282551 PMCID: PMC12025727 DOI: 10.3390/cancers17081375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths. The 5-year survival rate has been estimated to be less than 20% while its incidence rates have more than tripled since the 1980s. Astrocyte elevated gene-1/Metadherin (AEG-1/MTDH) has been demonstrated to have an influential role in HCC progression and the development of an aggressive phenotype. AEG-1 has been shown to be upregulated in many cancers, including HCC. Studies have shown that it plays a crucial role in the proliferation, invasion and metastasis, and evasion of apoptosis in HCC. Its relationship with proteins and pathways, such as MYC, SND1, PI3K/AKT, and other signaling pathways demonstrates its pertinent role in oncogenic development and relevance as a biomarker and therapeutic target. Recent studies have shown that AEG-1 is present in tumor tissues, and the anti-AEG-1 antibody is detected in the blood of cancer patients, demonstrating its viability as a diagnostic/prognostic marker. This review paper shines light on recent findings regarding the molecular implications of AEG-1, with emphasis on its role of regulating metabolic dysfunction-associated steatohepatitis (MASH), a key predisposing factor for HCC, new treatment strategies targeting AEG-1, and challenges associated with analyzing this intriguing molecule.
Collapse
Affiliation(s)
- Eva Davis
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Ali Gawi Ermi
- Department of Cellular, Molecular and Genetic Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Devanand Sarkar
- Department of Cellular, Molecular and Genetic Medicine, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
209
|
Liu JJ, Zhou M, Yuan T, Huang ZY, Zhang ZY. Conversion treatment for advanced intrahepatic cholangiocarcinoma: Opportunities and challenges. World J Gastroenterol 2025; 31:104901. [PMID: 40309227 PMCID: PMC12038554 DOI: 10.3748/wjg.v31.i15.104901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/22/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
The prevalence of intrahepatic cholangiocarcinoma (ICC) is increasing globally. Despite advancements in comprehending this intricate malignancy and formulating novel therapeutic approaches over the past few decades, the prognosis for ICC remains poor. Owing to the high degree of malignancy and insidious onset of ICC, numerous cases are detected at intermediate or advanced stages of the disease, hence eliminating the chance for surgical intervention. Moreover, because of the highly invasive characteristics of ICC, recurrence and metastasis postresection are prevalent, leading to a 5-year survival rate of only 20%-35% following surgery. In the past decade, different methods of treatment have been investigated, including transarterial chemoembolization, transarterial radioembolization, radiotherapy, systemic therapy, and combination therapies. For certain patients with advanced ICC, conversion treatment may be utilized to facilitate surgical resection and manage disease progression. This review summarizes the definition of downstaging conversion treatment and presents the clinical experience and evidence concerning conversion treatment for advanced ICC.
Collapse
Affiliation(s)
- Jun-Jie Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Mi Zhou
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Tong Yuan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zhi-Yong Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zun-Yi Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| |
Collapse
|
210
|
Sarsenbayeva A, Sadak S, Kucuk I, Kudreyeva L, Bakytzhanovna AM, Uslu B. Molybdenum-Based Electrochemical Sensors for Breast Cancer Biomarker Detection: Advances and Challenges. Crit Rev Anal Chem 2025:1-21. [PMID: 40257753 DOI: 10.1080/10408347.2025.2487581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Breast cancer, which is considered the most common type of cancer among women worldwide, is estimated to reach 4.4 million cases in 2070. Early diagnosis has become very important to prevent this expected increase. Various traditional methods, such as mammography, biopsy, enzyme immunoassay (EI), liquid biopsy, immunohistochemistry (IGH), fluorescence in situ hybridization (FISH) are used to diagnose breast cancer, but the fact that these methods are very expensive, have low sensitivity, and cause mutations in tissues due to X-rays has led researchers to discover faster, more cost-effective, and easily detectable methods. In particular, increased levels of new blood-based biomarkers in the circulation can be detected sensitively and selectively by electrochemical methods to facilitate early disease screening and rapid diagnosis. This comprehensive review focuses on the prevalence and pathology of breast cancer, clinical diagnosis of breast cancer, and electrochemical sensors of molybdenum-based compounds for the detection of various breast cancer biomarkers in recent years. Electrochemical analysis studies carried out in the field in recent years are compiled and are considered as aptamer-based, nucleotide-based, and immunosensors. The chemical properties of molybdenum compounds are discussed, and the modifications of these compounds to the electrode surface are discussed under 4 headings: drop casting, electrodeposition, atomic layer deposition, and electrophoretic deposition.
Collapse
Affiliation(s)
- Aliya Sarsenbayeva
- Department of Analytical Chemistry, Colloidal Chemistry and Technology of Rare Elements, Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Selenay Sadak
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
- The Graduate School of Health Sciences, Ankara University, Ankara, Turkey
| | - Ipek Kucuk
- The Graduate School of Health Sciences, Ankara University, Ankara, Turkey
- Department of Analytical Chemistry, Faculty of Pharmacy, Başkent University, Ankara, Turkey
| | - Leila Kudreyeva
- Department of Analytical Chemistry, Colloidal Chemistry and Technology of Rare Elements, Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Abu Moldir Bakytzhanovna
- Department of Analytical Chemistry, Colloidal Chemistry and Technology of Rare Elements, Faculty of Chemistry and Chemical Technology, Al-Farabi Kazakh National University, Almaty, Kazakhstan
| | - Bengi Uslu
- Department of Analytical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
211
|
Maurier L, Chéné AL, Hulo P, Chen J, Sagan C, Pons-Tostivint E. [Diffuse interstitial lung disease induced by antibody-drug conjugates]. Rev Mal Respir 2025:S0761-8425(25)00166-4. [PMID: 40263022 DOI: 10.1016/j.rmr.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 03/13/2025] [Indexed: 04/24/2025]
Abstract
INTRODUCTION Antibody-drug conjugates (ADCs) represent a promising new therapeutic class in non-small-cell lung cancer (NSCLC) patients. Studies assessing ADC have highlighted a pulmonary toxicity profile in the form of interstitial lung disease (ILD). STATE OF THE ART Several ADCs for NSCLC are currently being developed. In studies evaluating Trastuzumab-Deruxtecan (Her-2 target), incidence of drug-induced ILD ranged from 10.7 to 26.0%, and from 3.6 to 25.0% in those evaluating Datopotamab-Deruxtecan (TROP-2 target). Incidence of 9.9 and 5% of ILD was observed with Telisotuzumab-Vedotin (c-MET target) and Patritumab-Deruxtecan (Her-3 target), respectively. No cases of ILD have been reported with Sacituzumab-Govitecan (TROP-2 target) or Tusamitamab-Ravtansine (CEACAM5 target). PERSPECTIVES Several risk factors for ADC-induced ILD seem to emerge, including respiratory comorbidities, renal insufficiency, or type and dosage of ADC. Current studies are focusing on the combination of ADC and immunotherapy, although there are few data now available on pulmonary toxicity profiles. CONCLUSION Among the many ADCs being developed, several can cause ILD of varying grades and intensity. Knowledge of their risks, diagnostic and therapeutic modalities is required in order to quickly detect and treat ADC-induced ILD.
Collapse
Affiliation(s)
- L Maurier
- Service de pneumologie, hôpital Laennec, CHU de Nantes, boulevard Professeur-Jacques-Monod, 44800 Saint-Herblain, France
| | - A-L Chéné
- Service de pneumologie, hôpital Laennec, CHU de Nantes, boulevard Professeur-Jacques-Monod, 44800 Saint-Herblain, France; Service d'oncologie médicale, hôpital Laennec, CHU de Nantes, boulevard Professeur-Jacques-Monod, 44800 Saint-Herblain, France
| | - P Hulo
- Service d'oncologie médicale, hôpital Laennec, CHU de Nantes, boulevard Professeur-Jacques-Monod, 44800 Saint-Herblain, France
| | - J Chen
- Service d'oncologie médicale, hôpital Laennec, CHU de Nantes, boulevard Professeur-Jacques-Monod, 44800 Saint-Herblain, France
| | - C Sagan
- Service d'anatomopathologie, Hôtel-Dieu, CHU de Nantes, 1 place Alexis-Ricordeau, 44000 Nantes, France
| | - E Pons-Tostivint
- Service d'oncologie médicale, hôpital Laennec, CHU de Nantes, boulevard Professeur-Jacques-Monod, 44800 Saint-Herblain, France.
| |
Collapse
|
212
|
Shi P, Xu J, Cui H. Targeting oxygenases could be a viable anti-metastatic approach in cancer therapy. Int J Biol Macromol 2025; 310:143375. [PMID: 40268020 DOI: 10.1016/j.ijbiomac.2025.143375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
Malignant tumors are characterized by irregular boundaries, rapid and uncontrolled cell growth, the ability to invade surrounding tissues, and the potential to spread and metastasize to other parts of the body through the bloodstream or lymphatic system. More than 90 % of cancer-related deaths are attributed to the metastasis of cancer cells. When malignant tumors metastasize, the metabolic processes within the cells undergo significant changes, with enzymes playing a crucial role in regulating metabolism and serving as key mediators in both synthesis and degradation. Oxygenases are a group of oxidative enzymes that catalyze the incorporation of oxygen atoms into various substrates. Advances in our understanding of the genome and proteome of malignant tumors have revealed that oxygenases are highly expressed in many metastatic tumor cells, where they can enhance the activity of specific proteins that regulate tumor metastasis. Furthermore, there is a growing recognition that certain drugs can specifically target oxygenases to inhibit tumor metastasis, with several of these agents are currently undergoing clinical evaluation. In this context, we summarize the mechanisms by which oxygenases influence cancer cell behavior, along with the preclinical and clinical studies related to targeted therapies involving oxygenases.
Collapse
Affiliation(s)
- Pengfei Shi
- Jinfeng Laboratory, 401329 Chongqing, China; Cancer Center, Medical Research Institute, Southwest University, 400716 Chongqing, China
| | - Jie Xu
- Jinfeng Laboratory, 401329 Chongqing, China; Cancer Center, Medical Research Institute, Southwest University, 400716 Chongqing, China
| | - Hongjuan Cui
- Jinfeng Laboratory, 401329 Chongqing, China; Cancer Center, Medical Research Institute, Southwest University, 400716 Chongqing, China.
| |
Collapse
|
213
|
Önder T, Öner İ, Karaçin C, Ateş Ö. PIV and PILE scores predict the clinical outcome in patients with metastatic breast cancer treated with CDK4/6 inhibitors. Int J Clin Oncol 2025:10.1007/s10147-025-02770-w. [PMID: 40257656 DOI: 10.1007/s10147-025-02770-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
AIMS AND OBJECTIVES Cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) therapy, the standard of care for metastatic hormone receptor-positive (HR +)/human epidermal growth factor receptor 2 (HER2) negative breast cancer (BC), has profoundly affected many cell types, including tumor cells, Tregs, cytotoxic T cells, and stem and progenitor cells. Therefore, it is reasonable to assume that the pretreatment status of tumor immunity may have predictive value in CDK4/6i efficacy. METHODS A total of 404 patients were included in the analysis. The scores of the panimmune-inflammatory values (PIV) and PILE (PIV-LDH-ECOG), a candidate PIV-based scoring system, were calculated within one week before the initiation of CDK4/6i plus endocrine therapy (ET). RESULTS The median overall survival (OS) was 69.0 months (95% CI 51.1-86.8). The low-PIV subgroup had significantly longer progression-free survival (PFS) [23.9 vs. 18.8 months; HR = 1.817, 95% CI = 1.113-2.965, p = 0.017] and OS [73.6 vs. 37.7 months; HR = 2.338, 95% CI = 1.122-4.871, p = 0.023] than the high-PIV subgroup. In the low-risk PILE subgroup, PFS [37.0 vs. 15.8 months; HR = 2.751, 95% CI = 1.736-4.361, p < 0.001] and OS [73.6 vs. 35.1 months; HR = 3.854, 95% CI = 1.855-8.005, p < 0.001] were greater than in the high-risk PILE subgroup. The low-risk PILE subgroup was associated with a significantly better disease control rate (DCR) than the high-risk PILE subgroup (87.2% and 75.0%, p = 0.004). In the analysis of 112 patients treated with ET in the metastatic stage before CDK4/6i as a control group, PIV and PILE were not independent prognostic indicators. CONCLUSIONS Our study demonstrated that PIV and PILE scores could be predictive biomarkers for the treatment efficacy of CDK4/6is.
Collapse
Affiliation(s)
- Tuğba Önder
- Dr Abdurrahman Yurtaslan Ankara Oncology Education and Research Hospital, Health Sciences University, Ankara, Turkey.
| | - İrem Öner
- Dr Abdurrahman Yurtaslan Ankara Oncology Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Cengiz Karaçin
- Dr Abdurrahman Yurtaslan Ankara Oncology Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Öztürk Ateş
- Dr Abdurrahman Yurtaslan Ankara Oncology Education and Research Hospital, Health Sciences University, Ankara, Turkey
| |
Collapse
|
214
|
Li C, Fang L, Su X, Zhang J, Xiong H, Yu H, Zhu Z, Lin X, Min K, Wu D, Chen Z, Gong J, Xie CM. Macrophage miR-4524a-5p/TBP promotes β-TrCP -TIM3 complex activation and TGFβ release and aggravates NAFLD-associated fibrosis. Cell Death Dis 2025; 16:315. [PMID: 40251185 PMCID: PMC12008196 DOI: 10.1038/s41419-025-07574-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/20/2025]
Abstract
Macrophages hold a critical position in maintenance of hepatic homeostasis and in injury and repair processes in acute and chronic liver diseases. TIM3 is a promising protector in MCD-induced steatohepatitis in acute liver injury. However, we recently find TIM3 as a driver of fibrosis in MCD/HFD-induced chronic liver injury. This study aims to explore how macrophage TIM3 drivers NAFLD-associated chronic liver injury as well as identify a subtype of fibrotic patients suitable for anti-TIM3 immunotherapy. Here, we found that TIM3 was highly expressed in liver macrophages in a long-term MCD- or HFD-fed mice with fibrotic NASH. Elevated β-TrCP in macrophages promoted TIM3 polyubiquitination and membrane translocation. The ubiquitinated TIM3 then bound with PI3K and followed by inhibition of mTOR and activation of macrophage M2 polarization and TGF-β release, leading to HSC activation and liver fibrosis. Furthermore, elevated TIM3 was attributed to the transcriptional TBP upregulation and miR-4524a-5p downregulation. Targeting of TIM3 significantly attenuated liver fibrosis in mice. In clinical NASH patients, elevated macrophage TIM3 is positively correlated with TBP expression and negatively associated with miR-4524a-5p. Decreased miR-4524a-5p in plasma was a biomarker for the NASH fibrosis patients suitable for anti-TIM3 therapy. In conclusion, this study reveals that miR-4524a-5p/TBP promotes β-TrCP/TIM3 complex activation in macrophages and aggravates chronic NASH fibrosis, providing miR-4524a-5p as an effective blood biomarker for a subtype of chronic NASH patients with fibrosis suitable for anti-TIM3 treatment.
Collapse
Affiliation(s)
- Chunming Li
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Fang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xingxing Su
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jie Zhang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Haojun Xiong
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hongqiang Yu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhu Zhu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaotong Lin
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ke Min
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Di Wu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhiyu Chen
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Chuan-Ming Xie
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
215
|
Tsutsumi C, Ohuchida K, Imamura M, Tan B, Shimada Y, Son K, Kosai T, Katayama N, Mochida Y, Hayashida S, Iwamoto C, Torata N, Horioka K, Shindo K, Mizuuchi Y, Ikenaga N, Nakata K, Oda Y, Nakamura M. Prognostic nutrition index reveals LAG3 in cytotoxic CD8+ T cells and MHC class II in gastric cancer cells. Cancer Immunol Immunother 2025; 74:176. [PMID: 40252096 PMCID: PMC12009253 DOI: 10.1007/s00262-025-04037-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/25/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND The prognostic nutrition index (PNI) has recently been highlighted as a predictor of immune checkpoint (IC) inhibitor efficacy in gastric cancer (GC). Although LAG3, an IC molecule, has gained considerable attention, its association with PNI remains unexplored. MATERIALS AND METHODS We retrospectively analyzed clinical data from 796 GC patients who underwent radical gastrectomy to identify which previously reported nutritional index had the greatest impact on prognosis. Single-cell RNA sequencing was performed on 38 GC tissues, and multiplex immunofluorescence staining was conducted on 59 GC tissues to evaluate the relationship between nutritional indices and IC molecule expression in cytotoxic CD8-positive T cells. RESULTS A low preoperative PNI was identified as the strongest predictor of poor prognosis among the nutritional indices in GC patients. The expression of not only PDCD1 (encoding PD1) but also LAG3 in cytotoxic CD8-positive T cells was significantly higher in GC with low PNI compared to those with high PNI. Among cytotoxic CD8-positive T cells, the proportion of LAG3-positive cells was greater than that of PDCD1-positive cells, particularly in GC with low PNI, and most LAG3-positive cells did not co-express PDCD1. Additionally, the expression of MHC class II, a ligand for LAG3, was higher in GC cells with high levels of epithelial-mesenchymal transition-related molecules in GC with low PNI compared to those with high PNI. CONCLUSIONS PNI can reflect LAG3 expression in cytotoxic CD8-positive T cells and MHC class II expression in GC cells.
Collapse
Affiliation(s)
- Chikanori Tsutsumi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kenoki Ohuchida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Masaki Imamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Bryan Tan
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Shimada
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kiwa Son
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takaaki Kosai
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Naoki Katayama
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Mochida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Sayuri Hayashida
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Chika Iwamoto
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Nobuhiro Torata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kohei Horioka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Shindo
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yusuke Mizuuchi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Naoki Ikenaga
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kohei Nakata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
216
|
Li X, Lei T, Fu L, Gao R, Cao N, Gu Y, Su H, Guo T, Che Y. Meta-analysis of the efficacy of applying reduced surgery for the treatment of asymptomatic unresectable advanced gastric cancer. BMC Gastroenterol 2025; 25:271. [PMID: 40251493 PMCID: PMC12007127 DOI: 10.1186/s12876-025-03849-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/03/2025] [Indexed: 04/20/2025] Open
Abstract
OBJECTIVES Systematic evaluation of the efficacy and safety of reduction surgery in asymptomatic unresectable advanced gastric cancer. MATERIALS AND METHODS PubMed, EMBASE, Cochrane Library and Web of Science were searched from database inception to 12 July 2024. The Cochrane Risk of Bias Assessment Tool and Newcastle-Ottawa Scale were used to evaluate the quality and analyze the bias of the randomized controlled and non-randomized controlled studies included in this study, and RevMan (Version 5.4) was used to perform the meta-analysis. RESULTS A total of 5 studies were finally included, including 1 randomized controlled study and 4 retrospective studies. The cumulative sample size was 1717 cases, including 701 cases in the reduced surgery group and 1016 cases in the non-surgical treatment group. The results of the Meta-analysis showed that the reduced surgery group did not offer a survival benefit compared with the non-surgical treatment group in terms of 1-year, 3-year, and 5-year survival rates. The reduced surgery group had a longer median survival time than the non-surgical group by 11.58 months. The incidence rate, morbidity rate, and mortality rate of the reduced surgery group were 5.5% and 6.5% higher than those of the non-surgical group, respectively. The incidence of perioperative complications and death rate in the reduced surgery group were 15% and 4%, respectively; about 3% of patients might have complications of the primary foci during non-surgical treatment and need palliative surgical resection. CONCLUSION Current evidence suggests that in asymptomatic patients with unresectable advanced gastric cancer, reduced surgery with resection of the primary site does not result in a long-term survival benefit. We look forward to more high-quality randomized controlled trials to provide more substantial evidence to support clinical practice.
Collapse
Affiliation(s)
- Xiong Li
- Department Cadre Ward of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Ting Lei
- The First Hospital of Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Liangyin Fu
- Department Cadre Ward of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Ruiyu Gao
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Ning Cao
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Yuanhui Gu
- Department Cadre Ward of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - He Su
- Department Cadre Ward of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Tiankang Guo
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China
| | - Yang Che
- Department Cadre Ward of General Surgery, Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China.
- Gansu Provincial Hospital, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
217
|
Kano S, Kawakita D, Honma Y, Takahashi H, Nakaguro M, Utsumi Y, Saigusa N, Hanazawa T, Tsukahara K, Okada T, Okami K, Yamazaki K, Ueki Y, Saito Y, Ozawa H, Arai T, Shimizu A, Hanyu K, Iwaki S, Imaizumi S, Sakai A, Yamauchi M, Tanaka R, Sato Y, Yamamura K, Sekimizu M, Imanishi Y, Hirai H, Sato Y, Urano M, Yamamoto H, Fushimi C, Matsuki T, Nagao T, Tada Y. The impact of HER2-Low expression in salivary duct carcinoma: Clinicopathologic features, survival outcomes, and association with androgen receptor-targeted therapy. Oral Oncol 2025; 165:107280. [PMID: 40252453 DOI: 10.1016/j.oraloncology.2025.107280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 04/21/2025]
Abstract
OBJECTIVES Recent advances in systemic therapy for salivary duct carcinoma (SDC) have been driven by the development of HER2- and androgen receptor (AR)-targeted therapies. Trastuzumab deruxtecan has proven effective not only in HER2-positive but also HER2-low breast and gastro-esophageal cancers. However, the significance of HER2-low expression in SDC remains unknown. This study aimed to investigate the clinicopathologic characteristics, prognostic implications, and impact on efficacy to AR-targeted therapy in HER2-low SDC. MATERIALS AND METHODS This was a multi-center, observational study. HER2 status was reclassified as follows: HER2-positive (IHC3+ or 2+/ISH+ ), HER2-low (IHC1+ or 2+/ISH-), and HER2-zero (IHC0). The subjects were compared in three groups: total population, curative treatment cohort, and AR-targeted therapy cohort. RESULTS The total population consisted of 526 patients, of whom, 271 (52 %), 184 (35 %), and 71 (13 %) had HER2-positive, -low, and -zero tumors, respectively. Sex, M category, histological origin, Ki67, and p53 expression differed significantly between the HER2-low and HER2-positive cases. No differences in relapse-free or overall survival were observed for HER2 status in the curative treatment cohort; however, in the AR-targeted therapy cohort, the HER2-low group had significantly better response rates (41.6 % vs. 18.9 %, Odds ratio = 0.30, P = 0.012) and longer median progression-free survival (6.9 vs. 4.2 months, Hazard ratio = 1.61, P = 0.029) than those of the HER2-positive group. CONCLUSION HER2-low showed different clinicopathologic features from HER2-positive cases, with no prognostic differences observed in patients who underwent curative treatment. Still, HER2-low may be associated with the efficacy of AR-targeted therapy.
Collapse
Affiliation(s)
- Satoshi Kano
- Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Daisuke Kawakita
- Department of Otorhinolaryngology, Head and Neck Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoshitaka Honma
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hideaki Takahashi
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Masato Nakaguro
- Department of Pathology and Laboratory Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Yoshitaka Utsumi
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Natsuki Saigusa
- Division of Dental and Maxillofacial Radiology and Oral Pathology Diagnostic Services, The Nippon Dental University Hospital, Tokyo, Japan
| | - Toyoyuki Hanazawa
- Department of Otorhinolaryngology/Head & Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kiyoaki Tsukahara
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Takuro Okada
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University Hachioji Medical Center, Hachioji, Japan
| | - Kenji Okami
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Tokai University, Isehara, Japan
| | - Keisuke Yamazaki
- Department of Head and Neck Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Yushi Ueki
- Department of Otolaryngology Head and Neck Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yuki Saito
- Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Ozawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Tomoyuki Arai
- Department of Otorhinolaryngology/Head & Neck Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Akira Shimizu
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Kenji Hanyu
- Department of Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Sho Iwaki
- Department of Otorhinolaryngology, Head and Neck Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Sae Imaizumi
- Department of Otorhinolaryngology, Head and Neck Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akihiro Sakai
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Tokai University, Isehara, Japan
| | - Mayu Yamauchi
- Department of Otolaryngology, Head and Neck Surgery, School of Medicine, Tokai University, Isehara, Japan
| | - Ryoko Tanaka
- Department of Head and Neck Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Yuichiro Sato
- Department of Head and Neck Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Koji Yamamura
- Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mariko Sekimizu
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yorihisa Imanishi
- Otorhinolaryngology, Head and Neck Surgery, International University of Health and Welfare, Narita Hospital, Narita, Japan
| | - Hideaki Hirai
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Yukiko Sato
- Division of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makoto Urano
- Department of Diagnostic Pathology, Bantane Hospital, Fujita Health University, School of Medicine, Nagoya, Japan
| | - Hidetaka Yamamoto
- Pathology and Oncology, Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama University, Okayama, Japan
| | - Chihiro Fushimi
- Department of Head and Neck Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takashi Matsuki
- Department of Head and Neck Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Toshitaka Nagao
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Yuichiro Tada
- Department of Head and Neck Oncology and Surgery, International University of Health and Welfare, Mita Hospital, Tokyo, Japan.
| |
Collapse
|
218
|
Palaj J, Kečkéš Š, Marek V, Dyttert D, Sabol M, Durdík Š, Waczulíková I. Single centre experience with conversion surgery for advanced and metastatic gastric cancer in Slovakia. Sci Rep 2025; 15:13381. [PMID: 40251306 PMCID: PMC12008260 DOI: 10.1038/s41598-025-98656-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
Conversion surgery (CS) following systemic chemotherapy (SCT) has been suggested as a promising strategy for improving prognosis of patients with advanced gastric carcinoma (GC). The evidence, however, comes mostly from small-scale studies. Moreover, controversy exists over the criteria for selecting patients for SCT. We retrospectively analyzed 123 patients treated between 2007 and 2023. Thirty-one underwent CS, 44 received primary radical surgery (R0) with adjuvant chemotherapy (ACT), and 48 received surgical or palliative treatment. Survival rates and predictors of successful conversion were assessed. Median survival for R0 + ACT (30.4 months, 95%CI: 20.9-45.0) was non-significantly higher than SCT + R0 (19.4 months, 95%CI: 10.3-40.1; P = 0.2353). Successful downstaging after SCT was observed in 54.8% of CS patients. This group of SCT responders had significantly lower laboratory markers CEA, NLR and PLR (P-value of 0.019; 0.036 and 0.029, respectively). Both successful and failed conversion groups had significantly longer survival than group with palliative treatment (16.0 months, 95%CI: 8.4-19.1 vs. 7.4 months, 95%CI: 5.3-9.9; P = 0.0003). Multivariable analysis confirmed significantly lowered hazard and prolonged overall survival in CS vs. palliative treatments after adjusting for age and stage differences (P = 0.0014). Conversion therapy improves short-term survival and offers potential for long-term survival in select stage IV GC patients.
Collapse
Affiliation(s)
- Július Palaj
- Department of Surgical Oncology St. Elizabeth Cancer Institute, Heydukova 10, Bratislava, 812 50, Slovak Republic.
- Faculty of Medicine in Bratislava of the Comenius University, Špitálska 24, Bratislava, 813-72, Slovak Republic.
| | - Štefan Kečkéš
- Department of Immunodiagnostics, St. Elizabeth Cancer Institute, Bratislava, Slovak Republic
- St. Elizabeth University of Health and Social Sciences, Bratislava, Slovak Republic
| | - Víťezslav Marek
- Department of Surgical Oncology St. Elizabeth Cancer Institute, Heydukova 10, Bratislava, 812 50, Slovak Republic
- Faculty of Medicine in Bratislava of the Comenius University, Špitálska 24, Bratislava, 813-72, Slovak Republic
| | - Daniel Dyttert
- Department of Surgical Oncology St. Elizabeth Cancer Institute, Heydukova 10, Bratislava, 812 50, Slovak Republic
- Faculty of Medicine in Bratislava of the Comenius University, Špitálska 24, Bratislava, 813-72, Slovak Republic
| | - Martin Sabol
- Department of Surgical Oncology St. Elizabeth Cancer Institute, Heydukova 10, Bratislava, 812 50, Slovak Republic
- Faculty of Medicine in Bratislava of the Comenius University, Špitálska 24, Bratislava, 813-72, Slovak Republic
| | - Štefan Durdík
- Department of Surgical Oncology St. Elizabeth Cancer Institute, Heydukova 10, Bratislava, 812 50, Slovak Republic
- Faculty of Medicine in Bratislava of the Comenius University, Špitálska 24, Bratislava, 813-72, Slovak Republic
- St. Elizabeth University of Health and Social Sciences, Bratislava, Slovak Republic
| | - Iveta Waczulíková
- Department of Nuclear Physics and Biophysics, Division of Biomedical Physics, Faculty of Mathematics, Physics and Informatics, Comenius University, Bratislava, Slovak Republic
| |
Collapse
|
219
|
Bu S, Wang S, Wang T, Xing H, Cao Y, Zhang Z, Shang C, Tang X, Liu Y, Dong X, Wang X. Efficacy and safety of XELOX combined with neoadjuvant radiotherapy versus neoadjuvant chemotherapy in locally advanced gastric cancer. BMC Cancer 2025; 25:731. [PMID: 40251501 PMCID: PMC12007279 DOI: 10.1186/s12885-025-14103-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 04/07/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND The work aimed to compare the efficacy and safety of chemotherapy regimen (oxaliplatin + capecitabine, XELOX) combined with neoadjuvant radiotherapy (NART) and neoadjuvant chemotherapy (NACT) in locally advanced gastric cancer. METHODS We retrospectively analyzed clinical data from patients with locally advanced gastric cancer who underwent radical gastrectomy with D2 lymph node dissection at our center between January 2019 and December 2020. The study compared tumor markers, postoperative pathology, short-term efficacy, postoperative complications, and hospital stay between the chemoradiotherapy (CRT, XELOX + NART) group and the NACT-only group. Pearson correlation coefficients was used to analyze the correlations between clinical variables and tumor biomarkers. Inverse probability weighting (IPW) was used to adjust for confounding factors. RESULTS A total of 409 patients were included, with 369 (90.2%) in the NACT group and 40 (9.8%) in the CRT group. Significant correlations were found between clinical variables and tumor biomarkers, which may help identify potential prognostic factors for gastric cancer treatment. After IPW adjustment, baseline characteristics were similar between groups. The negative conversion rate of CEA-positive patients was significantly higher in the CRT group (38.1% vs. 11.8%, P < 0.001). The rate of pathological complete response was also higher in the CRT group (15.8% vs. 4.7%, P = 0.017). Postoperative pathological stages ypT0 and T1 were observed in 35.5% of the CRT group compared to 13.5% in the NACT group (P = 0.031). The CRT group had a lower average number of lymph nodes dissected (17 vs. 24, P < 0.001) but a higher ypN0 rate (60.3% vs. 39.8%, P = 0.024). The proportion of patients with tumor regression grade (TRG) 0-1 was higher in the CRT group (60.3% vs. 24.3%, P = 0.003). The R0 resection rate after IPW was 100% in the CRT group versus 96.5% in the NACT group (P = 0.001). No significant differences were found between the CRT and NACT groups in nerve invasion, vascular embolus, peritoneal invasion, bone marrow suppression, nausea, vomiting, esophagitis, diarrhea, other adverse reactions, postoperative complications, or average hospitalization time. The CRT group showed superior disease-free survival while no overall survival advantage (P < 0.05). CONCLUSIONS The XELOX regimen combined with neoadjuvant chemoradiotherapy provided superior downstaging, short-term pathological response, and local control benifits compared to perioperative chemotherapy alone, with similar surgical safety profiles.
Collapse
Affiliation(s)
- Shanshan Bu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Siyi Wang
- Department of Surgical Oncology and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ting Wang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Hang Xing
- Department of Surgery, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, USA
| | - Yue Cao
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, 22600, China
| | - Zhandong Zhang
- Department of General Surgery, The Affiliated Tumor Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Chuang Shang
- Department of General Surgery, The Affiliated Tumor Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Xiance Tang
- Department of Medical Record, The Affiliated Tumor Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, 22600, China.
| | - Xiaoqun Dong
- Precision Health Program, Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| | - Xiushen Wang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
220
|
Kaur R, Klassen PN, Mazurak VC. Improving analysis of sexual dimorphism in body composition dynamics in the oncology setting: A scoping review. Clin Nutr ESPEN 2025; 67:673-684. [PMID: 40254164 DOI: 10.1016/j.clnesp.2025.03.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/23/2025] [Accepted: 03/28/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND AND AIMS Chemotherapy treatments induce loss of skeletal muscle and adipose tissue each of which are important prognostic indicators after a cancer diagnosis. Males and females may respond differently to drugs used to treat cancer. Given the high degree of heterogeneity in the literature, the objective of this scoping review is to evaluate the methodological variability in reporting of muscle and adipose tissue changes comparing males and females during cancer-directed treatment. METHODS Relevant databases were searched for papers reporting longitudinal CT based body composition changes separately for males and females in solid tumors. RESULTS Of the 29 studies that met inclusion criteria, 22 were retrospective and 7 were prospective. The majority of studies reported on gastrointestinal cancers [n=24]. Among collective participants (n= 5139), 32% were females. Females were under represented in half the studies. For 21/29 studies, baseline characteristics were combined for males and females, hindering the ability to understand the effect of disease stage, chemotherapy type and co-morbidities on muscle and fat changes experienced by each sex. Multiple chemotherapy regimens were combined (n=24) and not reported in a sex-specific way (n=26). CONCLUSION While the literature reporting body composition changes during cancer treatment is abundant, study design and reporting is problematic and precludes metaanalysis. Disproportionate numbers of males and females, marked heterogeneity in cancer types and chemotherapy regimens evaluated within a single study collectively pose challenges in analysing the impact of specific chemotherapy regimens on muscle and adipose change by sex. Strategies to standardize this set of literature in a sex specific way are required to improve evidence synthesis.
Collapse
Affiliation(s)
- Ravneet Kaur
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Pamela N Klassen
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Vera C Mazurak
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
221
|
Liu X, Zhang J, Yi T, Li H, Tang X, Liu D, Wu D, Li Y. Decoding tumor angiogenesis: pathways, mechanisms, and future directions in anti-cancer strategies. Biomark Res 2025; 13:62. [PMID: 40251641 PMCID: PMC12007322 DOI: 10.1186/s40364-025-00779-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/13/2025] [Indexed: 04/20/2025] Open
Abstract
Angiogenesis, a crucial process in tumor growth and metastasis, necessitates targeted therapeutic intervention. This review reviews the latest knowledge of anti-angiogenesis targets in tumors, with emphasis on the molecular mechanisms and signaling pathways that regulate this process. We emphasize the tumor microenvironment's role in angiogenesis, examine endothelial cell metabolic changes, and evaluated potential therapeutic strategies targeting the tumor vascular system. At the same time, we analyzed the signaling pathway and molecular mechanism of tumor angiogenesis in detail. In addition, this paper also looks at the development trend of tumor anti-angiogenesis drugs, including their future development direction and challenges, aiming to provide prospective insight into the development of this field. Despite their potential, anti-angiogenic therapies encounter challenges like drug resistance and side effects, necessitating ongoing research to enhance cancer treatment strategies and the efficacy of these therapies.
Collapse
Affiliation(s)
- Xueru Liu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Ting Yi
- Department of Trauma Center, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Xing Tang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Dan Liu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Daichao Wu
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China.
| |
Collapse
|
222
|
付 玮, 宁 静, 付 伟, 张 静, 丁 士. [Effect of CMTM6 on PD-L1 in Helicobacter pylori infected gastric epithelial cells]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2025; 57:245-252. [PMID: 40219552 PMCID: PMC11992445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Indexed: 04/14/2025]
Abstract
OBJECTIVE To explore the changes of CKLF-like MARVEL transmembrane domain-containing 6 (CMTM6) and programmed death-ligand 1 (PD-L1) expression in gastric mucosal epithelial cells after Helicobacter pylori infection and the regulation of CMTM6 on PD-L1, and to analyze the mRNA expression differences before and after CMTM6 gene knock-out in helicobacter pylori infected gastric epithelial cells by microarray analysis. METHODS The standard Helicobacter pylori strain ATCC 26695 was co-cultured with human gastric epithelial cell GES-1 for 6, 24 and 48 hours, and the mRNA and protein levels of CMTM6 and PD-L1 were detected by real-time quantitative PCR and Western blot. Using CRISPR/Cas9 to construct CMTM6 gene knockout plasmid and knockout CMTM6 gene of GES-1 cells. Helicobacter pylori was co-cultured with CMTM6 gene knockout and wild type GES-1 cells for 48 hours to detect PD-L1 transcription and protein level changes, and CMTM6 gene knockout GES-1 cells were treated with the proteasome inhibitor MG-132 to detect the changes in PD-L1 protein levels. Agilent Human ceRNA Microarray 2019 was used to detect the differentially expressed genes in CMTM6 gene knockout and wild-type GES-1 cells co-cultured with Hp for 48 hours, and the signal pathway of differentially expressed genes enrichment was analyzed by Kyoto Encyclopedia of Genes and Genomes (KEGG) database. RESULTS The mRNA and protein levels of CMTM6 and PD-L1 in GES-1 cells were significantly up-regulated after Helicobacter pylori infection, and CMTM6 mRNA was most significantly up-regulated 48 hours after infection. After CMTM6 gene knockout, the CD274 gene transcription level of Helicobacter pylori infected GES-1 cells did not change significantly, but PD-L1 protein level was significantly down-regulated, and the PD-L1 level increased after the application of proteasome inhibitor MG-132. After CMTM6 gene knockout, 67 genes had more than two times of differential expression. The transcription levels of TMEM68, FERMT3, GPR142, ATP6V1FNB, NOV, UBE2S and other genes were significantly down-regulated. The transcription levels of PCDHGA6, CAMKMT, PDIA2, NTRK3, SPOCK1 and other genes were significantly up-regulated. After CMTM6 gene knockout, ubiquitin-conjugating enzyme E2S (UBE2S) gene expression was significantly down-regulated, which might affect protein ubiquitination degradation. After CMTM6 gene knockout, adrenoceptor alpha 1B (ADRA1B), cholinergic receptor muscarinic 1 (M1), CHRM1, platelet activating factor receptor (PTAFR) gene expression was significantly up-regulated. CONCLUSION Helicobacter pylori infection up-regulates the expression level of CMTM6 in gastric mucosa cells, and CMTM6 can stabilize PD-L1 and maintain the protein level of PD-L1. CMTM6 gene knockout may affect biological behaviors such as protein ubiquitination and cell surface receptor expression.
Collapse
Affiliation(s)
- 玮 付
- />北京大学第三医院消化科,幽门螺杆菌感染及上胃肠疾病防治研究北京市重点实验室,北京 100191Department of Gastroenterology, Peking University Third Hospital; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - 静 宁
- />北京大学第三医院消化科,幽门螺杆菌感染及上胃肠疾病防治研究北京市重点实验室,北京 100191Department of Gastroenterology, Peking University Third Hospital; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - 伟伟 付
- />北京大学第三医院消化科,幽门螺杆菌感染及上胃肠疾病防治研究北京市重点实验室,北京 100191Department of Gastroenterology, Peking University Third Hospital; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - 静 张
- />北京大学第三医院消化科,幽门螺杆菌感染及上胃肠疾病防治研究北京市重点实验室,北京 100191Department of Gastroenterology, Peking University Third Hospital; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - 士刚 丁
- />北京大学第三医院消化科,幽门螺杆菌感染及上胃肠疾病防治研究北京市重点实验室,北京 100191Department of Gastroenterology, Peking University Third Hospital; Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| |
Collapse
|
223
|
QUAN JINGDAN, WAN ZIXIN, WU WEI, CAO XINYUAN, QIU JIAYUAN, LIU XIAOYE, ZHANG ZHIWEI. Classical biomarkers and non-coding RNAs associated with diagnosis and treatment in gastric cancer. Oncol Res 2025; 33:1069-1089. [PMID: 40296904 PMCID: PMC12034007 DOI: 10.32604/or.2025.063005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
One of the most prevalent malignant tumors worldwide, stomach cancer still has a high incidence and fatality rate in China, and the number of young people developing early-onset gastric cancer is steadily increasing. The 5-year survival rate of stomach cancer is typically 30%-35%, the prognosis is bad, the patients' quality of life is low, and the progression of advanced gastric cancer cannot be effectively managed despite the use of surgical surgery, chemotherapy, and other medicines. We urgently need molecular biomarkers with high specificity and sensitivity to increase the early gastric cancer detection rate, extend patient survival, and improve patient quality of life. The initial diagnosis of gastric cancer primarily depends on gastroscopy and biopsy, and invasive procedures cause significant discomfort to patients. Similar to this, treating advanced and metastatic stomach cancer is a pressing issue that requires attention. More and more immune checkpoint molecules have been discovered, and corresponding inhibitors are gradually being applied to clinical diagnosis and treatment. Recently, some non-coding RNAs have begun to be used as new targets for the treatment of gastric cancer. Some non-coding RNAs are highly present in the serum or urine of gastric cancer patients and can be used as diagnostic markers or prognostic indicators. Many clinical trials targeting non-coding RNAs have also shown good therapeutic effects. In general, targeting non-coding RNAs has shown good therapeutic effects. The biomarkers for gastric cancer detection and treatment are reviewed in this article, focusing on the new non-coding RNAs used in diagnosis, prognosis, and treatment. Patients with stomach cancer should have access to more precise and efficient diagnosis and treatment choices as a result of ongoing technological advancements and thorough research.
Collapse
Affiliation(s)
- JINGDAN QUAN
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - ZIXIN WAN
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - WEI WU
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - XINYUAN CAO
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - JIAYUAN QIU
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - XIAOYE LIU
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - ZHIWEI ZHANG
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, 421001, China
| |
Collapse
|
224
|
Stenzinger A, Westphalen CB, Budczies J, Kazdal D, Ploeger C, Altbürger C, Evert M, Malek N, Schirmacher P, Klauschen F. Comprehensive genomic profiling requires a blended ecosystem of learning healthcare and clinical trials. Br J Cancer 2025:10.1038/s41416-025-03009-1. [PMID: 40251279 DOI: 10.1038/s41416-025-03009-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/03/2025] [Accepted: 03/28/2025] [Indexed: 04/20/2025] Open
Affiliation(s)
- Albrecht Stenzinger
- Center for Personalized Medicine (ZPM), Heidelberg, Germany.
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
| | - C Benedikt Westphalen
- Comprehensive Cancer Center and Department of Medicine III, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jan Budczies
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Daniel Kazdal
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carolin Ploeger
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Altbürger
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Nisar Malek
- Center for Personalized Medicine (ZPM), Tübingen, Germany
- Department for Internal Medicine 1, Tübingen University Hospital, Tübingen, Germany
| | - Peter Schirmacher
- Center for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Frederick Klauschen
- Institute of Pathology, Ludwig Maximilian University of Munich, Munich, Germany
- BIFOLD-Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| |
Collapse
|
225
|
Li X, Mendez Q, Chapados C, Acca F, Driscoll H, Oliveira J, Liu J, Jones K, Ferguson M, Wallace RL, Bibikov S, Lionberger T, Harvey KJ, Weiner MP, Mirando G. Site-directed antibodies targeting driver mutations of the KRAS protein. N Biotechnol 2025; 87:112-120. [PMID: 40252917 DOI: 10.1016/j.nbt.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/14/2025] [Accepted: 04/05/2025] [Indexed: 04/21/2025]
Abstract
Kirsten rat sarcoma viral oncogene homolog (KRAS) is the most mutated oncogene in human cancers, found in approximately 30 % of tumors. These mutations primarily consist of single-base missense alterations in codon G12. While extensive efforts have focused on developing allele-specific inhibitors for KRAS mutations, mutation-specific antibodies (Abs) remain largely unexplored, with only a few research-use-only catalog Abs available. In this study, we employed the proprietary Epivolve technology to develop site-directed monoclonal Abs (mAbs) that target KRAS oncogenic driver mutation KRAS G12D. These site-directed mAbs demonstrate high binding affinity, with equilibrium dissociation constants (KD) in the nanomolar range, showing over 1,000-fold greater affinity for KRAS G12D compared to wild-type KRAS. Western blot analyses using both purified KRAS protein variants and tumor cell lines harboring G12D mutations confirmed the high specificity of these mAbs. Furthermore, immunocytochemistry analysis revealed co-localization of the site-directed mAbs with endogenously expressed KRAS in cancer cells bearing G12D mutations. The validated high affinity and specificity of these site-directed mAbs highlight their potential for diagnostic applications and therapeutic development targeting KRAS driver mutations.
Collapse
Affiliation(s)
- Xiaofeng Li
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA.
| | - Qiana Mendez
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | | | - Felicity Acca
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Holly Driscoll
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Jason Oliveira
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Jun Liu
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Kezzia Jones
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Mary Ferguson
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Ryan L Wallace
- Aviva Systems Biology Corporation, 6370 Nancy Ridge Dr., Suite 104, San Diego, CA 92121, USA
| | - Sergei Bibikov
- Aviva Systems Biology Corporation, 6370 Nancy Ridge Dr., Suite 104, San Diego, CA 92121, USA
| | - Troy Lionberger
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Kevin J Harvey
- Aviva Systems Biology Corporation, 6370 Nancy Ridge Dr., Suite 104, San Diego, CA 92121, USA
| | - Michael P Weiner
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Greg Mirando
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| |
Collapse
|
226
|
Peng Z, Zhang X, Liang H, Zheng Z, Wang Z, Liu H, Hu J, Sun Y, Zhang Y, Yan H, Tong L, Xu J, Ji J, Shen L. Atezolizumab and Trastuzumab Plus Chemotherapy for ERBB2-Positive Locally Advanced Resectable Gastric Cancer: A Randomized Clinical Trial. JAMA Oncol 2025:2832721. [PMID: 40244574 PMCID: PMC12006909 DOI: 10.1001/jamaoncol.2025.0522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/10/2025] [Indexed: 04/18/2025]
Abstract
Importance Effective treatment of locally advanced gastric cancer (GC) or gastroesophageal junction (GEJ) cancer remains a challenge. Objective To compare the efficacy and safety of atezolizumab plus trastuzumab plus capecitabine and oxaliplatin chemotherapy (XELOX) vs trastuzumab plus XELOX in Chinese patients with locally advanced human epidermal growth factor receptor 2 (ERBB2; formerly HER2)-positive GC or adenocarcinoma of the GEJ. Design, Setting, and Participants This was an open-label phase 2 randomized clinical trial conducted at 8 study sites in China. Patient recruitment started on February 25, 2021, and this study is ongoing as participants are still being actively followed up. Chinese patients eligible for surgery with locally advanced ERBB2-positive GC or adenocarcinoma of the GEJ were included. Data were analyzed from March 2021 to October 2023. Interventions Eligible patients were enrolled and randomly assigned 1:1 to perioperative treatment with either atezolizumab plus trastuzumab plus XELOX (arm A) or trastuzumab plus XELOX (arm B) for 3 neoadjuvant cycles (3 weeks per cycle) and 5 adjuvant cycles. Main Outcomes and Measures The primary efficacy end point was the pathological complete response (pCR) rate following completion of neoadjuvant therapy and surgery. Results In total, 42 patients were screened and randomly assigned to arm A (n = 21) or arm B (n = 21). The median (range) ages were 61 (33-72) years and 65 (49-72) years in arm A and arm B, respectively, and 39 patients (93%) were male. The pCR rate was significantly higher in arm A (8 [38%]) than arm B (3 [14%]; difference, 23.8%; 90% CI, 1.3-44.7). Age younger than 65 years, male sex, and intestinal Lauren classification were significantly associated with a better pCR rate in arm A. Median event-free survival, disease-free survival, and overall survival were not reached. Based on the same way of interpretation, major pathologic response should be statistically significantly different between the 2 arms, while other outcome measures remained not significantly different. The incidence of treatment-emergent adverse events was 100% (21 of 21) and 100% (21 of 21) in arms A and B, respectively; grade 3 or higher TEAEs, 57% (12 of 21) and 67% (14 of 21), respectively; and serious TEAEs, 29% (6 of 21) and 10% (2 of 21), respectively. Conclusions and Relevance In this randomized clinical trial, add-on atezolizumab to trastuzumab plus XELOX therapy demonstrated promising efficacy in this patient population, and no new safety concerns were raised. Trial Registration ClinicalTrials.gov Identifier: NCT04661150.
Collapse
Affiliation(s)
- Zhi Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Xiaotian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Han Liang
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | | | - Zhenning Wang
- Department of Surgical Oncology, First Hospital of China Medical University, Shenyang, China
| | - Hao Liu
- General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiankun Hu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yihong Sun
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yanqiao Zhang
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Han Yan
- Shanghai Roche Pharmaceuticals, Shanghai, China
| | - Lin Tong
- Shanghai Roche Pharmaceuticals, Shanghai, China
| | - Jiahui Xu
- Shanghai Roche Pharmaceuticals, Shanghai, China
| | - Jiafu Ji
- Peking University Cancer Hospital and Institute, Beijing, China
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
227
|
Fathy A, Allam A, ElHady AK, El-Gamil DS, Lin KC, Chang YH, Lee YH, Hilscher S, Schutkowski M, Ibrahim HS, Chen SH, Chen CH, Abadi AH, Sippl W, Chen PJ, Cheng YS, Abdel-Halim M. Development of potent and selective tetrahydro-β-carboline-based HDAC6 inhibitors with promising activity against triple-negative breast cancer. RSC Med Chem 2025:d5md00086f. [PMID: 40256307 PMCID: PMC12004265 DOI: 10.1039/d5md00086f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/26/2025] [Indexed: 04/22/2025] Open
Abstract
Overexpression of histone deacetylase 6 (HDAC6) is implicated in tumorigenesis, invasion, migration, survival, apoptosis, and growth of various malignancies, making it a promising target for cancer treatment. Building on our previous work, we report a novel series of tetrahydro-β-carboline-piperazinedione derivatives as HDAC6 inhibitors. Structural modifications were introduced at the 6-aryl group, with the m-bromophenyl derivative (9c) emerging as the most potent HDAC6 inhibitor, exhibiting an IC50 of 7 nM. Compound 9c demonstrated robust growth inhibitory activity across 60 cancer cell lines from the NCI panel, with a mean GI50 of 2.64 μM and a GI50 below 5 μM for nearly all tested lines, while exhibiting significantly lower cytotoxicity towards non-tumor cell lines. The triple-negative breast cancer cell line MDA-MB-231 was selected for further investigation of 9c's cellular effects. 9c selectively increased the acetylation of non-histone α-tubulin in MDA-MB-231 cells, confirming its HDAC6 selectivity. Furthermore, 9c effectively induced apoptosis, caused apoptotic sub-G1 phase accumulation, upregulated pro-apoptotic caspase-3, and downregulated anti-apoptotic Bcl-2. Notably, 9c reduced the expression of programmed death-ligand 1 (PD-L1), a key immune checkpoint protein that enables tumor cells to evade immune surveillance, highlighting its potential role in enhancing anti-tumor immunity. In addition, 9c inhibited phosphorylated extracellular signal-regulated kinase (ERK)1/2, a central signaling pathway that drives cell proliferation, survival, and migration, further highlighting its significance in suppressing tumor progression and growth. In migration assays, 9c impaired cell motility, achieving 80% gap closure inhibition in a wound-healing assay. Collectively, these findings underline compound 9c as a highly promising candidate for the treatment of triple-negative breast cancer, with the added benefits of PD-L1 and ERK inhibition for potential synergy in enhancing anti-tumor immunity and reducing tumor cell proliferation.
Collapse
Affiliation(s)
- Aya Fathy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo Cairo 11835 Egypt
| | - Amro Allam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo Cairo 11835 Egypt
| | - Ahmed K ElHady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo Cairo 11835 Egypt
- School of Life & Medical Sciences, University of Hertfordshire hosted by Global Academic Foundation New Administrative Capital Cairo Egypt
| | - Dalia S El-Gamil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo Cairo 11835 Egypt
- Department of Chemistry, Faculty of Pharmacy, Ahram Canadian University Cairo 12451 Egypt
| | - Kai-Chun Lin
- Institute of Plant Biology, College of Life Science, National Taiwan University Taipei 10617 Taiwan
| | - Yen-Hua Chang
- Institute of Plant Biology, College of Life Science, National Taiwan University Taipei 10617 Taiwan
| | - Yu-Hsuan Lee
- Department of Life Science, College of Life Science, National Taiwan University Taipei 10617 Taiwan
| | - Sebastian Hilscher
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg Halle (Saale) Germany
| | - Mike Schutkowski
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg Halle (Saale) Germany
| | - Hany S Ibrahim
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg Halle (Saale) Germany
| | - Shun-Hua Chen
- School of Nursing, Fooyin University Kaohsiung 831301 Taiwan
| | - Chun-Hong Chen
- Department of Medical Research, E-Da Hospital and Graduate Institute of Medicine, I-Shou University Kaohsiung 824410 Taiwan
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo Cairo 11835 Egypt
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther-University of Halle-Wittenberg Halle (Saale) Germany
| | - Po-Jen Chen
- Department of Medical Research, E-Da Hospital and Graduate Institute of Medicine, I-Shou University Kaohsiung 824410 Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Kaohsiung Medical University Kaohsiung 80708 Taiwan
| | - Yi-Sheng Cheng
- Institute of Plant Biology, College of Life Science, National Taiwan University Taipei 10617 Taiwan
- Department of Life Science, College of Life Science, National Taiwan University Taipei 10617 Taiwan
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University Taipei 10617 Taiwan
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo Cairo 11835 Egypt
| |
Collapse
|
228
|
Li J, Tao M, Liu L, Liu C, Ma M, Liu D, Zhang P, Zhang M, Xue R, Gong J, Zhang C, Zhang X, Shen L, Qi C. Peripheral blood neutrophils contribute to Claudin18.2-specific CAR-T cell treatment resistance in advanced gastric cancer. Br J Cancer 2025:10.1038/s41416-025-03015-3. [PMID: 40246985 DOI: 10.1038/s41416-025-03015-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Claudin18.2 (CLDN18.2)-specific chimeric antigen receptor (CAR)-T cell treatment holds promise for advanced gastric cancer (GC) but has variable efficacy. This study investigates the prognostic value of the neutrophil-to-lymphocyte ratio (NLR) in CAR-T cell treatment and elucidates the molecular mechanisms of treatment resistance. METHODS GC patients treated with CLDN18.2-specific CAR-T cell treatment were analyzed. Outcomes included objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). Survival analyses utilized Kaplan-Meier methods, log-rank tests, and Cox regression. Single-cell RNA sequencing was performed on peripheral blood samples to investigate the mechanisms of pro-tumor circulating neutrophils. RESULTS Elevated NLR was significantly associated with lower ORR (34.2% vs. 55.9%, P < 0.001), shorter median PFS (3.6 vs. 8.0 months, P < 0.001), and OS (5.6 vs. 13.8 months, P < 0.001). Single-cell sequencing identified a circulating neutrophil subcluster (NE-3) linked to disease progression. NE-3 expressed pro-tumoral factors (MMP-9), and was enriched in the IL-17 signaling pathway. The cellular interactions between neutrophils and T cells were more prominent in progression disease (PD) group than in partial response (PR) group. CONCLUSIONS This study highlights NLR as a significant prognostic factor in advanced GC patients receiving CLDN18.2-specific CAR-T cell treatment and provides insights into neutrophil-mediated treatment resistance. Further validation and exploration of strategies to mitigate neutrophil-induced immunosuppression are needed. TRIAL REGISTRATION NCT03874897.
Collapse
Affiliation(s)
- Jiarui Li
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Min Tao
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lian Liu
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Chang Liu
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Mingyang Ma
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Dan Liu
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Panpan Zhang
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Miao Zhang
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ran Xue
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jifang Gong
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Cheng Zhang
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaotian Zhang
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Lin Shen
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Changsong Qi
- Beijing Key Laboratory of Cell & Gene Therapy for Solid Tumor, State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
229
|
Theik NWY, De Armas SA, Rosas D, Kiamos A, Thaw Dar NN, Shoreibah A, Hussein A, Raez LE. Oncogenic Fusions in NSCLC: From Mechanisms to Clinical Applications. Int J Mol Sci 2025; 26:3802. [PMID: 40332427 DOI: 10.3390/ijms26083802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/17/2025] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) is operated commonly by diverse genetic alterations, and oncogenic fusions represent a significant therapeutic role. Common fusions include ALK, ROS1, RET, and NTRK, signaling pathways in tumorigenesis. Recent advances in investigating tumor molecular biology include underlying fusions, including chromosomal rearrangements, highlighting their role as oncogenic drivers. The development of targeted therapies, such as tyrosine kinase inhibitors (TKIs), has impacted most patients' NSCLC treatment. Despite the greater profiles, such as remarkable efficiency and tolerable side effects compared to traditional chemotherapy, challenges, such as acquired mutations, lead to more ongoing research-optimized future NSCLC therapies.
Collapse
Affiliation(s)
- Nyein Wint Yee Theik
- Memorial Healthcare System, Internal Medicine Residency Program, Pembroke Pines, FL 33028, USA
| | - Suset Almuinas De Armas
- Memorial Healthcare System, Internal Medicine Residency Program, Pembroke Pines, FL 33028, USA
| | - Daniel Rosas
- Memorial Cancer Institute, Memorial Healthcare System, Hematology-Oncology Fellowship Program, Pembroke Pines, FL 33028, USA
| | - Amy Kiamos
- Memorial Cancer Institute, Memorial Healthcare System, Hematology-Oncology Fellowship Program, Pembroke Pines, FL 33028, USA
| | - Nyein Nyein Thaw Dar
- Memorial Cancer Institute, Memorial Healthcare System, Hematology-Oncology Fellowship Program, Pembroke Pines, FL 33028, USA
| | - Ahmed Shoreibah
- Memorial Healthcare System, Internal Medicine Residency Program, Pembroke Pines, FL 33028, USA
| | - Atif Hussein
- Memorial Cancer Institute, Memorial Healthcare System, Florida Atlantic University (FAU), Hollywood, FL 33021, USA
| | - Luis E Raez
- Thoracic Oncology Program, Memorial Cancer Institute (MCI), Florida Atlantic University (FAU), Hollywood, FL 33021, USA
| |
Collapse
|
230
|
Yousef M, Yousef A, Hurd MW, Pillai A, Chowdhury S, Snyder R, Knafl M, Lewis RL, Roy PM, Fanaeian M, Albarouki S, Castelnovo LF, Peterson J, Smaglo BG, Wolff RA, Pant S, Willis J, Huey R, Overman M, Tzeng CW, Kim MP, Ikoma N, Maxwell JE, Katz MHG, Wang H, Maitra A, Koay E, Ludmir EB, Chen A, Lopez C, Ying H, Shen JP, Zhao D. KRAS mutation detection by liquid biopsy for pancreatic ductal adenocarcinoma. J Hematol Oncol 2025; 18:44. [PMID: 40247392 PMCID: PMC12004821 DOI: 10.1186/s13045-025-01696-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/02/2025] [Indexed: 04/19/2025] Open
Abstract
The clinical utility of liquid biopsy (LB) for pancreatic ductal adenocarcinoma (PDAC) remain understudied. Our single-institution cohort of 311 PDAC patients with non-tumor tissues informed LB found 81.2% positivity (N = 186) in metastatic cases and in 52.4% (N = 43) of localized disease. KRAS mutations were detected in 64.6% (N = 148) of metastatic cases and 16% (N = 13) for localized disease. Positive LB, especially KRAS mutation detection, is associated with worse overall survival (OS) in metastatic PDAC (median 14.5 vs. 31.3 months, HR = 2.7, 95%CI = 1.7-4.3, P < 0.0001). The positive concordance rates of KRAS and TP53 mutations were 63% and 68% in metastatic disease but only 7% (KRAS) and 33% (TP53) in localized disease, respectively. Among the 41 patients who underwent serial liquid biopsy testing, 25% tested positive after an initial negative result. LB detects therapeutically targetable mutations in 58.5% of PDAC patients and is associated with OS.
Collapse
Affiliation(s)
- Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark W Hurd
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ashwathy Pillai
- Department of Hospital Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rebecca Snyder
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark Knafl
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ryan L Lewis
- Department of Enterprise Data Engineering and Analytics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul M Roy
- Department of Enterprise Data Engineering and Analytics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohammad Fanaeian
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sali Albarouki
- Department of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - Luca F Castelnovo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer Peterson
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brandon G Smaglo
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jason Willis
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ryan Huey
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ching-Wei Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael P Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jess E Maxwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew H G Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Huamin Wang
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene Koay
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ethan B Ludmir
- Department of Gastrointestinal Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anthony Chen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Camila Lopez
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, Division of Basic Science Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dan Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
231
|
Yuan T, Liu Y, Wu R, Qian M, Wang W, Li Y, Zhu H, Wang J, Ge F, Zeng C, Dai X, Hu R, Zhou T, He Q, Zhu H, Yang B. Josephin Domain Containing 2 (JOSD2) inhibition as Pan-KRAS-mutation-targeting strategy for colorectal cancer. Nat Commun 2025; 16:3623. [PMID: 40240366 PMCID: PMC12003847 DOI: 10.1038/s41467-025-58923-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
KRAS is the most common mutated oncogenes in colorectal cancer (CRC), yet effective therapeutic strategies for targeting multiple KRAS mutations remained challenging. The prolonged protein stability of KRAS mutants contribute to their robust tumor-promoting effects, but the underlying mechanism is elusive. Herein by screening deubiquitinases (DUBs) siRNA library, we identify Josephin domain containing 2 (JOSD2) functions as a potent DUB that regulates the protein stability of KRAS mutants. Mechanistically, JOSD2 directly interacts with and stabilizes KRAS variants across different mutants, by reverting their proteolytic ubiquitination; while KRAS mutants reciprocally inhibit the catalytic activity of CHIP, a bona fide E3 ubiquitin ligase for JOSD2, thus forming a JOSD2/KRAS positive feedback circuit that significantly accelerates KRAS-mutant CRC growth. Inhibition of JOSD2 by RNA interference or its pharmacological inhibitor promotes the polyubiquitination and proteasomal degradation of KRAS mutants, and preferentially impede the growth of KRAS-mutant CRC including patient-derived cells/xenografts/organoids (PDCs/PDXs/PDOs) over that harboring wild-type KRAS. Collectively, this study not only reveals the crucial roles of JOSD2/KRAS positive feedback circuit in KRAS-mutant CRC, but also provides a rationale to target JOSD2 as the promising pan-KRAS-mutation-targeting strategy for the treatment of a broad population of CRC patients with KRAS variant across different mutant types.
Collapse
Affiliation(s)
- Tao Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yue Liu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ruilin Wu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Meijia Qian
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Weihua Wang
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yonghao Li
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hongdao Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jia'er Wang
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Fujing Ge
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chenming Zeng
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyang Dai
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, China
| | - Ronggui Hu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianhua Zhou
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, China
| | - Hong Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- School of Medicine, Hangzhou City University, Hangzhou, China.
| |
Collapse
|
232
|
Leivaditis V, Maniatopoulos AA, Lausberg H, Mulita F, Papatriantafyllou A, Liolis E, Beltsios E, Adamou A, Kontodimopoulos N, Dahm M. Artificial Intelligence in Thoracic Surgery: A Review Bridging Innovation and Clinical Practice for the Next Generation of Surgical Care. J Clin Med 2025; 14:2729. [PMID: 40283559 PMCID: PMC12027631 DOI: 10.3390/jcm14082729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/27/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Artificial intelligence (AI) is rapidly transforming thoracic surgery by enhancing diagnostic accuracy, surgical precision, intraoperative guidance, and postoperative management. AI-driven technologies, including machine learning (ML), deep learning, computer vision, and robotic-assisted surgery, have the potential to optimize clinical workflows and improve patient outcomes. However, challenges such as data integration, ethical concerns, and regulatory barriers must be addressed to ensure AI's safe and effective implementation. This review aims to analyze the current applications, benefits, limitations, and future directions of AI in thoracic surgery. Methods: This review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A comprehensive literature search was performed using PubMed, Scopus, Web of Science, and Cochrane Library for studies published up to January 2025. Relevant articles were selected based on predefined inclusion and exclusion criteria, focusing on AI applications in thoracic surgery, including diagnostics, robotic-assisted surgery, intraoperative guidance, and postoperative care. A risk of bias assessment was conducted using the Cochrane Risk of Bias Tool and ROBINS-I for non-randomized studies. Results: Out of 279 identified studies, 36 met the inclusion criteria for qualitative synthesis, highlighting AI's growing role in diagnostic accuracy, surgical precision, intraoperative guidance, and postoperative care in thoracic surgery. AI-driven imaging analysis and radiomics have improved pulmonary nodule detection, lung cancer classification, and lymph node metastasis prediction, while robotic-assisted thoracic surgery (RATS) has enhanced surgical accuracy, reduced operative times, and improved recovery rates. Intraoperatively, AI-powered image-guided navigation, augmented reality (AR), and real-time decision-support systems have optimized surgical planning and safety. Postoperatively, AI-driven predictive models and wearable monitoring devices have enabled early complication detection and improved patient follow-up. However, challenges remain, including algorithmic biases, a lack of multicenter validation, high implementation costs, and ethical concerns regarding data security and clinical accountability. Despite these limitations, AI has shown significant potential to enhance surgical outcomes, requiring further research and standardized validation for widespread adoption. Conclusions: AI is poised to revolutionize thoracic surgery by enhancing decision-making, improving patient outcomes, and optimizing surgical workflows. However, widespread adoption requires addressing key limitations through multicenter validation studies, standardized AI frameworks, and ethical AI governance. Future research should focus on digital twin technology, federated learning, and explainable AI (XAI) to improve AI interpretability, reliability, and accessibility. With continued advancements and responsible integration, AI will play a pivotal role in shaping the next generation of precision thoracic surgery.
Collapse
Affiliation(s)
- Vasileios Leivaditis
- Department of Cardiothoracic and Vascular Surgery, Westpfalz Klinikum, 67655 Kaiserslautern, Germany; (V.L.); (H.L.); (A.P.); (M.D.)
| | | | - Henning Lausberg
- Department of Cardiothoracic and Vascular Surgery, Westpfalz Klinikum, 67655 Kaiserslautern, Germany; (V.L.); (H.L.); (A.P.); (M.D.)
| | - Francesk Mulita
- Department of General Surgery, General Hospital of Eastern Achaia—Unit of Aigio, 25100 Aigio, Greece
| | - Athanasios Papatriantafyllou
- Department of Cardiothoracic and Vascular Surgery, Westpfalz Klinikum, 67655 Kaiserslautern, Germany; (V.L.); (H.L.); (A.P.); (M.D.)
| | - Elias Liolis
- Department of Oncology, General University Hospital of Patras, 26504 Patras, Greece;
| | - Eleftherios Beltsios
- Department of Anesthesiology and Intensive Care, Hannover Medical School, 30625 Hannover, Germany;
| | - Antonis Adamou
- Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, 30625 Hannover, Germany;
| | - Nikolaos Kontodimopoulos
- Department of Economics and Sustainable Development, Harokopio University, 17676 Athens, Greece;
| | - Manfred Dahm
- Department of Cardiothoracic and Vascular Surgery, Westpfalz Klinikum, 67655 Kaiserslautern, Germany; (V.L.); (H.L.); (A.P.); (M.D.)
| |
Collapse
|
233
|
Imaoka H, Ikeda M, Ozaka M, Oshima K, Okano N, Shimizu S, Tsumura H, Komatsu Y, Yamashita T, Kataoka S, Nagano H, Hisano T, Sasaki M, Kobayashi S, Fukushima T, Mitsunaga S, Furukawa T, Hamauchi S, Ueno M, Furuse J. Phase 1/2 study of liposomal irinotecan plus S-1 for metastatic pancreatic cancer refractory to gemcitabine-based treatment. Eur J Cancer 2025; 222:115424. [PMID: 40252631 DOI: 10.1016/j.ejca.2025.115424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Liposomal irinotecan (nal-IRI) plus fluorouracil/folinic acid (5-FU/LV) improves survival in gemcitabine-refractory metastatic pancreatic cancer (PC) but requires a central venous port. S-1, an oral fluoropyrimidine with proven efficacy in PC, may replace 5-FU/LV in nal-IRI plus 5-FU/LV, potentially enhancing both convenience and antitumor effect. METHODS This single-arm, open-label, phase 1/2 study included patients with histologically or cytologically confirmed adenocarcinoma, aged 20-80 years, an Eastern Cooperative Oncology Group performance status of 0-1, with metastatic disease, and refractory to gemcitabine-based treatment. The primary endpoint in phase 1 part was the frequency of dose-limiting toxicity (DLT) to nal-IRI plus S-1. The primary endpoint in phase 2 part was overall survival. This trial was registered in the Japan Registry of Clinical Trials database (jRCTs031210040). RESULTS In phase 1 part, one patient with DLT was observed at nal-IRI 70 mg/m2 (day 1) with S-1 80 mg/m2/day (day 1-7) in a 2-week cycle, establishing this as the recommended phase 2 dose (RP2D). Forty-nine patients from phase 1 (n = 6) and phase 2 part (n = 43) were treated with the RP2D, and their results were pooled. Median overall survival was 10.3 months (95 % confidence interval, 8.1-12.0 months). A confirmed partial response was achieved in 10 patients (20.4 %). The most frequent treatment-emergent adverse events were hypoalbuminemia (98.0 %), anemia (98.0 %), and anorexia (81.6 %). There were no treatment-related deaths. CONCLUSIONS This study demonstrated that nal-IRI plus S-1 exhibited promising efficacy and an acceptable safety profile in patients with metastatic PC refractory to gemcitabine-based treatment.
Collapse
Affiliation(s)
- Hiroshi Imaoka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masato Ozaka
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kotoe Oshima
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Satoshi Shimizu
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Hidetaka Tsumura
- Department of Gastroenterological Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Yoshito Komatsu
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa, Japan
| | - Shigeki Kataoka
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Terumasa Hisano
- Department of Hepato-Biliary-Pancreatology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Mitsuhito Sasaki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Satoshi Kobayashi
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Taito Fukushima
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Shuichi Mitsunaga
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takaaki Furukawa
- Department of Hepato-Biliary-Pancreatic Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoshi Hamauchi
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Junji Furuse
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
234
|
Kafeel S, Palmiero G, Salzillo A, Ragone A, Naviglio S, Sapio L. Combining AdipoRon with Paclitaxel Unveils Synergistic Potential in Non-Small Cell Lung Cancer Cells via AMPK-ERK1/2 Signaling. Cells 2025; 14:602. [PMID: 40277927 PMCID: PMC12026066 DOI: 10.3390/cells14080602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/02/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
As part of chemotherapy regimens, Paclitaxel improves the overall survival of many non-small cell lung cancer (NSCLC) patients. However, the development of drug resistance and adverse events limits its clinical usage, reinforcing the need for further advancements in NSCLC therapeutics. We recently recognized the adiponectin receptor agonist AdipoRon as a promising anticancer compound in NSCLC. Consequently, this study aimed to evaluate the therapeutic potential of combining AdipoRon with Paclitaxel (Combo) in NSCLC cells. With respect to individual treatments, Combo triggered a stronger inhibition of both cell growth and clonogenic potential, as well as a greater induction of cell death. The Combo-mediated cytotoxicity was also corroborated by cleavage of poly-ADP ribose polymerase (PARP) and caspase-3 apoptotic markers. Notably, AMP-activated protein kinase (AMPK) emerged as a critical sensor in Combo efficacy, as its inhibition by Compound-C unveiled a significant rescue in cell growth. Although Combo caused a gradual downregulation of extracellular signal-regulated kinase 1/2 (ERK1/2), the hindrance in the upstream cascade by PD98059 partially counteracted the Combo outcomes. In conclusion, our findings designate AdipoRon as an effective candidate in Paclitaxel-based therapy. Nevertheless, future studies aimed at exploring the Combo aptitude in overcoming the Paclitaxel-related restraints need to be investigated in NSCLC.
Collapse
Affiliation(s)
- Sanober Kafeel
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.K.); (G.P.); (A.S.); or (A.R.); (L.S.)
| | - Giuseppina Palmiero
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.K.); (G.P.); (A.S.); or (A.R.); (L.S.)
| | - Alessia Salzillo
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.K.); (G.P.); (A.S.); or (A.R.); (L.S.)
| | - Angela Ragone
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.K.); (G.P.); (A.S.); or (A.R.); (L.S.)
- Department of Mechanistic Cell Biology, Max Plank Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Silvio Naviglio
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.K.); (G.P.); (A.S.); or (A.R.); (L.S.)
| | - Luigi Sapio
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (S.K.); (G.P.); (A.S.); or (A.R.); (L.S.)
| |
Collapse
|
235
|
Mechahougui H, Gutmans J, Gouasmi R, Smekens L, Friedlaender A. BRAF Targeting Across Solid Tumors: Molecular Aspects and Clinical Applications. Int J Mol Sci 2025; 26:3757. [PMID: 40332392 DOI: 10.3390/ijms26083757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
BRAF mutations are critical drivers in cancers such as melanoma, colorectal cancer, and non-small-cell lung cancer. The most common mutation, BRAF V600E, is a key therapeutic target. Targeted treatments with BRAF and MEK inhibitors have significantly improved progression-free and overall survival in melanoma patients. However, in cancers like metastatic colorectal cancer, BRAF mutations are associated with poor outcomes due to aggressive disease behavior and resistance to conventional chemotherapy. Despite progress, resistance to BRAF/MEK inhibitors remains a major challenge, often driven by secondary mutations in the mitogen-activated protein kinase (MAPK) pathway, activation of alternative pathways such as phosphoinositide 3-kinases (PI3Ks)/protein kinase B (AKT), or changes in the tumor microenvironment. These challenges have motivated ongoing research into combining BRAF inhibitors with immunotherapies to enhance and prolong treatment effectiveness. Future research must also account for the role of the cancer's tissue of origin, as the biological context significantly influences response to targeted therapies, highlighting the need for a deeper understanding of tumor biology, micro-environment, and genetics.
Collapse
Affiliation(s)
- Hiba Mechahougui
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland
| | - James Gutmans
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland
| | - Roumaïssa Gouasmi
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, 69100 Lyon, France
| | - Laure Smekens
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland
| | | |
Collapse
|
236
|
Jawad RAM, Mshimesh BAR, Al-Mayah QS, Al-Alloosh F. A Case Study on Complete Pathological Response in Advanced Rectal Cancer Patient with Oxaliplatin-based Chemotherapy without Cumulative Neurotoxicity. J Gastrointest Cancer 2025; 56:99. [PMID: 40240738 DOI: 10.1007/s12029-025-01227-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND The pathological response in rectal cancer treatment provides insight into the molecular mechanisms, including genetic alterations and signaling pathways that influence tumor behavior and resistance to treatment. CASE PRESENTATION This report describes a 34-year-old Iraqi male diagnosed with stage III rectal cancer who achieved a complete pathological response following treatment with oxaliplatin-based chemotherapy. Notably, this outcome was achieved without the administration of chemoradiotherapy or the occurrence of neurotoxicity despite the efficacious cumulative‑dose administration (1700 mg/m2) of oxaliplatin. Genomic analysis revealed the presence of a heterozygous (Ile/Val) genotype in the GSTP1 gene, which may have contributed to the observed treatment response. CONCLUSIONS Genetic biomarkers play a crucial role in refining treatment strategies by enabling a more precise selection of patients who may safely forgo radiotherapy, thereby minimizing its associated toxicities. Additionally, molecular profiling can help predict susceptibility to oxaliplatin-induced neurotoxicity, facilitating dose adjustments or alternative therapeutic approaches to enhance treatment tolerance and long-term quality of life. Our findings highlight the importance of molecular profiling in optimizing treatment strategies while minimizing toxicity, especially in situations where radiological assessments suggest residual disease or produce unclear results.
Collapse
Affiliation(s)
- Rehab A M Jawad
- Department of Pharmacology and Toxicology, College of Pharmacy, Mustansiriyah University, Baghdad, Iraq.
- Ministry of Health, Kimadia, Baghdad, Iraq.
| | | | - Qasim S Al-Mayah
- Medical Research Unit, College of Medicine, Al-Nahrain University, Baghdad, Iraq
| | | |
Collapse
|
237
|
Zhang J, Zhu H, Liu W, Miao J, Mao Y, Li Q. Prognostic and predictive molecular biomarkers in colorectal cancer. Front Oncol 2025; 15:1532924. [PMID: 40308511 PMCID: PMC12040681 DOI: 10.3389/fonc.2025.1532924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Precision medicine has brought revolutionary changes to the diagnosis and treatment of cancer patients, and is currently a hot and challenging research topic. Currently, the treatment regimens for most colorectal cancer (CRC) patients are mainly determined by several biomakers, including Microsatellite Instability (MSI), RAS, and BRAF. However, the roles of promising biomarkers such as HER-2, consensus molecular subtypes (CMS), and circulating tumor DNA (ctDNA) in CRC are not yet fully clear. Therefore, it is urgent to explore the potential of these emerging biomarkers in the diagnosis and treatment of CRC patients. In this paper, we discuss recent advances in CRC biomarkers, especially clinical data, and focus on the roles of biomarkers in prognosis, prediction, treatment strategies, and the intrinsic connections with clinical pathological features, hoping to promote better precision medicine for colorectal cancer.
Collapse
Affiliation(s)
- Jianzhi Zhang
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Hao Zhu
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wentao Liu
- Department of General Surgery, Affiliated Drum Tower Hospital, JiangSu University, Nanjing, China
| | - Ji Miao
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yonghuan Mao
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Qiang Li
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
238
|
Wickramasinghe C, Kim S, Jiang Y, Bao X, Yue Y, Jiang J, Hong A, Sanai N, Li J. Population Pharmacokinetic Modeling of Total and Unbound Pamiparib in Glioblastoma Patients: Insights into Drug Disposition and Dosing Optimization. Pharmaceutics 2025; 17:524. [PMID: 40284519 PMCID: PMC12030534 DOI: 10.3390/pharmaceutics17040524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Background: This study aimed to develop a population pharmacokinetic (PK) model that characterized the plasma concentration-time profiles of the total and unbound pamiparib, a PARP inhibitor, in glioblastoma patients and identified patient factors influencing the PK. Methods: The total and unbound pamiparib plasma concentration data were obtained from 41 glioblastoma patients receiving 60 mg of pamiparib twice daily. Nonlinear mixed-effects modeling was performed using Monolix (2024R1) to simultaneously fit the total and unbound drug plasma concentration data. The covariate model was developed by covariate screening using generalized additive modeling followed by stepwise covariate modeling. Model simulations were performed following oral doses of 10-60 mg BID. Results: The total and unbound pamiparib plasma concentration-time profiles were best described by a one-compartment model with first-order absorption and elimination. Creatinine clearance and age were the significant covariates on the apparent volume of distribution (V/F) and apparent clearance (CL/F), respectively, explaining ~22% and ~5% of IIV of V/F and CL/F. Population estimates of the absorption rate constant (Ka), V/F, CL/F, and unbound fraction for the total drug were 1.58 h-1, 44 L, 2.59 L/h, and 0.041. Model simulations suggested that doses as low as 20 mg BID may be adequate for therapeutic effects in a general patient population, assuming that a target engagement ratio (i.e., unbound Css,min/IC50) of 5 or above is sufficient for full target engagement. Conclusions: The total and unbound pamiparib plasma PK are well characterized by a linear one-compartment model, with creatinine clearance as the significant covariate on V/F. Model simulations support further clinical investigation into dose reduction to optimize the benefit-to-risk ratio of pamiparib, particularly in combination therapies.
Collapse
Affiliation(s)
- Charuka Wickramasinghe
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Seongho Kim
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yuanyuan Jiang
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xun Bao
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yang Yue
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jun Jiang
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Amy Hong
- Barrow Neurological Institute, St. Joseph’s Hospital & Medical Center, Phoenix, AZ 85013, USA
| | - Nader Sanai
- Barrow Neurological Institute, St. Joseph’s Hospital & Medical Center, Phoenix, AZ 85013, USA
| | - Jing Li
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
239
|
Kim YR, Bae K, Lee JY, Jeong SW, Yoon HY, Han HJ, Hyun JE, Nam A, Park JH, Yoon KA, Kim JH. Clinical Utility of Patient-Derived Cell-Based In Vitro Drug Sensitivity Testing for Optimizing Adjuvant Therapy in Dogs with Solid Tumors: A Retrospective Study (2019-2023). Animals (Basel) 2025; 15:1146. [PMID: 40281980 PMCID: PMC12023965 DOI: 10.3390/ani15081146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/02/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
Inter-individual variations in drug responses are major concerns in cancer treatment in human and veterinary oncology. Consequently, preclinical models have been proposed to predict drug responses and determine optimal individualized therapy. We aimed to evaluate the clinical utility of in vitro drug sensitivity testing using a patient-derived cell culture model to select appropriate adjuvant therapies for dogs with solid tumors. We screened medical records of 126 dogs with suspected tumors, including 33 dogs with solid tumors (guided group, 16; empirical group, 17). Anticancer drugs used for adjuvant therapy were determined based on in vitro drug sensitivity testing (guided group) or histopathological examination (empirical group) results. Time to tumor progression (TTP) was compared between groups. The guided group had significantly longer TTP than the empirical group (949 vs. 109 days). Median TTPs were significantly longer in the guided group than in the empirical group for dogs with incomplete surgical margin (949 vs. 109 days), dogs with mitotic count < 20 per 10 high power fields (949 vs. 105 days), dogs with no evidence of metastatic disease at initial diagnosis (455 vs. 196 days), and dogs receiving tyrosine kinase inhibitors (949 vs. 109 days). Our study suggests that in vitro drug sensitivity testing may be a useful tool for optimizing adjuvant therapy in dogs with solid tumors.
Collapse
Affiliation(s)
- Young-Rok Kim
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul 05029, Republic of Korea
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Kieun Bae
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul 05029, Republic of Korea
- Department of Veterinary Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Ja-Young Lee
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul 05029, Republic of Korea
- Department of Veterinary Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Soon-Wuk Jeong
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul 05029, Republic of Korea
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hun-Young Yoon
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul 05029, Republic of Korea
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyun-Jung Han
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul 05029, Republic of Korea
- Department of Veterinary Emergency and Critical Care, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jae-Eun Hyun
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul 05029, Republic of Korea
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Aryung Nam
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul 05029, Republic of Korea
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Ji-Hwan Park
- Bundang Leaders Animal Medical Center, Seongnam 13636, Republic of Korea
| | - Kyong-Ah Yoon
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul 05029, Republic of Korea
- Department of Veterinary Biochemistry, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Jung-Hyun Kim
- KU Animal Cancer Center, Konkuk University Veterinary Medical Teaching Hospital, Seoul 05029, Republic of Korea
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
240
|
Peng S, Long M, Chen Q, Yin Z, Zeng C, Zhang W, Wen Q, Zhang X, Ke W, Wu Y. Perspectives on cancer therapy-synthetic lethal precision medicine strategies, molecular mechanisms, therapeutic targets and current technical challenges. Cell Death Discov 2025; 11:179. [PMID: 40240755 PMCID: PMC12003663 DOI: 10.1038/s41420-025-02418-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/27/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
In recent years, synthetic lethality has become an important theme in the field of targeted cancer therapy. Synthetic lethality refers to simultaneous defects in two or more genes leading to cell death, whereas defects in any single gene do not lead to cell death. Taking advantage of the genetic vulnerability that exists within cancer cells, it theoretically has no negative impact on healthy cells and has fewer side effects than non-specific chemotherapy. Currently, targeted cancer therapies focus on inhibiting key pathways in cancer. However, it has been found that over-activation of oncogenic-related signaling pathways can also induce cancer cell death, which is a major breakthrough in the new field of targeted therapies. In this review, we summarize the conventional gene targets in synthetic lethality (PARP, ATR, ATM, WEE1, PRMT) and provide an in-depth analysis of their latest potential mechanisms. We explore the impact of over-activation of pathways such as PI3K/AKT, MAPK, and WNT on cancer cell survival, and present the technical challenges of current research. Important theoretical foundations and insights are provided for the application of synthetic lethal strategies in cancer therapy, as well as future research directions.
Collapse
Affiliation(s)
- Shixuan Peng
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Mengle Long
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Qisheng Chen
- Department of Anesthesiology, The First People's Hospital of Chenzhou, The Chenzhou Affiliated Hospital, Hengyang Medical School, University of South China, Chenzhou, Hunan, 423000, China
| | - Zhijian Yin
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Chang Zeng
- Department of Pathology, Yueyang Central Hospital, Yueyang, China
| | - Wanyong Zhang
- Department of Pathology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, 437100, Hubei, China
| | - Qingyang Wen
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Xinwen Zhang
- Department of Oncology, Graduate Collaborative Training Base of The First People's Hospital of Xiangtan City, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Department of Oncology, The First People's Hospital of Xiangtan City, Xiangtan, Hunan, 411101, China
| | - Weiqi Ke
- Department of Anesthesiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China.
| | - Yongjun Wu
- Department of Pathology, Xiangtan Center Hospital, Xiangtan City, Hunan province, 411100, China.
- Department of Pathology, The Affiliated Hospital of Hunan University, Xiangtan City, Hunan Province, China.
| |
Collapse
|
241
|
Sreenivasan S, Jiwani RA, White R, Bakalov V, Moll R, Liput J, Greenberg L. Advances in Targeted and Systemic Therapy for Salivary Gland Carcinomas: Current Options and Future Directions. Curr Oncol 2025; 32:232. [PMID: 40277788 PMCID: PMC12025620 DOI: 10.3390/curroncol32040232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
Salivary gland carcinomas (SGCs) represent a rare and heterogeneous group of malignancies accounting for 3-6% of all head and neck cancers. While surgical resection and radiotherapy remain the standard for locoregional control, systemic treatment is indicated for recurrent or metastatic disease. Advances in molecular profiling have identified actionable targets such as NTRK gene fusions, HER2, immune checkpoint regulators, androgen receptors, and RET receptors. These have facilitated the development of targeted therapies, including TRK inhibitors, HER2-directed agents, and androgen receptor modulators, as well as emerging combinations of immunotherapy and chemotherapy. Despite these advancements, challenges such as resistance mechanisms and limited therapeutic efficacy persist. Overall response rates remain relatively low across most systemic therapies, reflecting a persistent unmet clinical need. This review discusses the current landscape of treatment options and explores promising clinical trials and future directions to enhance outcomes for patients with SGCs.
Collapse
Affiliation(s)
- Sushanth Sreenivasan
- Division of Internal Medicine, Allegheny Health Network, 320 East North Ave, Pittsburgh, PA 15212, USA
| | - Rahim A. Jiwani
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| | - Richard White
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| | - Veli Bakalov
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| | - Ryan Moll
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| | - Joseph Liput
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| | - Larisa Greenberg
- Division of Medical Oncology, Allegheny Health Network, 314 East North Ave, Pittsburgh, PA 15212, USA (V.B.)
| |
Collapse
|
242
|
He WZ, Yang YZ, Yin CX, Xian XY, Gu JM, Yi JH, Xue J, Zhao Y, Wang F, Hu WM, Xia LP. Evolution of HER2-low expression from primary to paired metastatic gastric cancer lesions. NPJ Precis Oncol 2025; 9:108. [PMID: 40234621 PMCID: PMC12000306 DOI: 10.1038/s41698-025-00881-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/15/2025] [Indexed: 04/17/2025] Open
Abstract
HER2-low expression has recently gained considerable attention as an actionable biomarker in gastric cancer. However, changes in HER2-low expression between primary and metastatic gastric cancers remain inadequately explored. This study included consecutive patients diagnosed with metastatic gastric cancer with both primary and metastatic tumors, between January 2014 and December 2023. HER2 status was evaluated in both primary and matched metastatic tumors. A total of 332 patients were enrolled, with HER2-negative, HER2-low, and HER2-positive statuses were observed in 226, 81, and 25 primary tumors, respectively, and in 175, 104, and 53 metastatic tumors, respectively. Among the 226 patients with HER2-negative primary tumors, 74 and 23 developed HER2-low and HER2-positive metastatic tumors, respectively. Conversion from HER2-negative primary to HER2-low metastatic gastric cancer was associated with metachronous and non-peritoneal metastasis. Overall, re-biopsy to evaluate HER2 status may be necessary, potentially broadening the patient population eligible for targeted HER2 therapy.
Collapse
Affiliation(s)
- Wen-Zhuo He
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Yuan-Zhong Yang
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Chen-Xi Yin
- Department of Intensive Care Unit, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Xin-Yi Xian
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Jia-Mei Gu
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Jia-Hong Yi
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Ju Xue
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Yue Zhao
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Fang Wang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| | - Wan-Ming Hu
- Department of Pathology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| | - Liang-Ping Xia
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China.
| |
Collapse
|
243
|
Su Y, Sun J, Li X, Huang F, Kong Y, Chen Z, Zhang J, Qin D, Chen X, Wang Z, Pei Y, Gong M, Yang K, Xu M, Dong Y, He Q, Zhang ZN, Sheng Z, Deng Q, Wang H, Wang G, Hu P, Le R, Gao S, Li W. CD47-blocking antibody confers metabolic benefits against obesity. Cell Rep Med 2025:102089. [PMID: 40267910 DOI: 10.1016/j.xcrm.2025.102089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 07/22/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025]
Abstract
CD47-blocking antibody is a well-known potential antibody drug for tumor immunotherapy. However, it is unclear whether CD47-blocking antibody can protect against metabolic disorders. We report that high-fat diet (HFD)-induced obesity increases CD47 expression, while exercise downregulates it in skeletal muscle. Administration of CD47-blocking antibody in mice prevents HFD-induced weight gain and glucose intolerance, enhances exercise capacity, and improves body composition and skeletal muscle mitochondrial function. Mechanistically, the protective effects conferred by CD47-blocking antibody are mediated through activation of AMP-activated protein kinase (AMPK) in skeletal muscle. Consistently, muscle-specific CD47-knockout mice show similar metabolic improvements, indicating a direct muscle-specific role of CD47 in regulating AMPK activation in vivo. Furthermore, the CD47-blocking antibody reduces the phosphorylation of heat shock protein 90α (HSP90α) to activate AMPK in skeletal muscle. In conclusion, CD47-blocking antibody confers metabolic benefits by activating the AMPK pathway in skeletal muscle.
Collapse
Affiliation(s)
- Yajuan Su
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China; Tsingtao Advanced Research Institute, Tongji University, Qingdao 266071, China
| | - Jingyu Sun
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Xiaobo Li
- Tsingtao Advanced Research Institute, Tongji University, Qingdao 266071, China
| | - Feier Huang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Yunhui Kong
- Institute of Modern Biology, Nanjing University, Nanjing 210023, China
| | - Zian Chen
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Jingzhi Zhang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Duran Qin
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Xiangyi Chen
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Zhaoyue Wang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Yu Pei
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Solna, Sweden
| | - Mengting Gong
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Kaijiang Yang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Minglu Xu
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Yu Dong
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Qing He
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Zhen-Ning Zhang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Zhejin Sheng
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institute, 17177 Solna, Sweden
| | - Hong Wang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China
| | - Gaowei Wang
- Institute of Modern Biology, Nanjing University, Nanjing 210023, China
| | - Ping Hu
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai TenthPeople's Hospital Affiliated to Tongji University, Shanghai 200031, China; Guangzhou Laboratory, No. 9 XingDaoHuan Road, Guanghzou International Bio lsland, Guangzhou 510005, China
| | - Rongrong Le
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China.
| | - Shaorong Gao
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China; Tsingtao Advanced Research Institute, Tongji University, Qingdao 266071, China.
| | - Weida Li
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Sports and Health Research Center, Tongji University Department of Physical Education, Tongji University, Shanghai 200092, China; Tsingtao Advanced Research Institute, Tongji University, Qingdao 266071, China.
| |
Collapse
|
244
|
Zhang SH, Li W, Chen XY, Nie LL. Combining immune checkpoint inhibitors with standard treatment regimens in advanced human epidermal growth factor receptor-2 positive gastric cancer patients. World J Gastrointest Oncol 2025; 17:103855. [PMID: 40235908 PMCID: PMC11995313 DOI: 10.4251/wjgo.v17.i4.103855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/12/2025] [Accepted: 02/07/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Gastric cancer is one of the most common malignant tumors worldwide, with its incidence and mortality rates ranking among the highest in gastrointestinal cancers. The overexpression or gene amplification of human epidermal growth factor receptor 2 (HER-2) occurs in approximately 15%-20% of gastric cancers and serves as a critical molecular target influencing prognosis and treatment outcomes. For patients with HER-2-positive gastric cancer, trastuzumab combined with platinum-based chemotherapy has been established as the standard first-line treatment. However, despite the demonstrated clinical benefits in prolonging survival, the overall efficacy remains limited. In recent years, with the successful application of immune checkpoint inhibitors (ICIs) in various malignant tumors, combining ICIs with existing standard treatment regimens has emerged as a promising approach to enhance the therapeutic efficacy of HER-2-positive gastric cancer. Nevertheless, the efficacy and prognostic factors of ICIs combined with trastuzumab and chemotherapy in HER-2-positive gastric cancer remain unclear. AIM To analyze the efficacy of ICIs combined with standard treatment regimens and the prognostic factors in patients with advanced HER-2-positive gastric cancer. METHODS Clinical data from 104 patients with advanced HER-2-positive gastric cancer who were treated at our hospital between March 2021 and May 2023 were retrospectively analyzed. Patients were divided into a control group (n = 54, treated with trastuzumab combined with platinum-based chemotherapy as the standard regimen) and an observation group (n = 50, treated with ICIs in addition to the standard regimen). The therapeutic efficacy, survival outcomes, and adverse reactions were compared between the two groups. Univariate and Cox multivariate analyses were performed to identify factors influencing patient prognosis. RESULTS With a median follow-up time of 14.6 months, there were no significant differences between the two groups in terms of objective response rate or disease control rate (P > 0.05). The median progression-free survival (mPFS) and mPFS for patients with immunohistochemistry 3 + in the observation group were significantly higher than those in the control group (P < 0.05). Among patients in the observation group, those with positive programmed death-ligand 1 (PD-L1) expression had a significantly higher mPFS than those with negative PD-L1 expression (P < 0.05). Regarding adverse events, significant differences were observed between the two groups in hypothyroidism and neutropenia (P < 0.05). Cox multivariate analysis showed that Eastern Cooperative Oncology Group (ECOG) performance status, peritoneal metastasis, positive programmed death-1 expression, and treatment regimen were independent factors influencing PFS (hazard ratio > 1, P < 0.05). CONCLUSION ICIs combined with standard treatment regimens for patients with advanced HER-2-positive gastric cancer demonstrate favorable clinical efficacy, significantly prolonging PFS with manageable safety. ECOG performance status, peritoneal metastasis, positive PD-L1 expression, and treatment regimen are independent factors influencing PFS, warranting increased clinical attention to patients exhibiting these factors.
Collapse
Affiliation(s)
- Sheng-Hu Zhang
- Department of Oncology, Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze university, Jingzhou 434020, Hubei Province, China
| | - Wan Li
- Department of Ultrasound Medicine, Jingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze university, Jingzhou 434020, Hubei Province, China
| | - Xi-Yan Chen
- Department of Medicine Imaging, The First People’s Hospital of Fuzhou City, Fuzhou 344000, Jiangxi Province, China
| | - Le-Le Nie
- Department of General Surgery, The First People’s Hospital of Fuzhou City, Fuzhou 344000, Jiangxi Province, China
| |
Collapse
|
245
|
Chen Y, Jia K, Xie Y, Yuan J, Liu D, Jiang L, Peng H, Zhong J, Li J, Zhang X, Shen L. The current landscape of gastric cancer and gastroesophageal junction cancer diagnosis and treatment in China: a comprehensive nationwide cohort analysis. J Hematol Oncol 2025; 18:42. [PMID: 40234884 PMCID: PMC12001465 DOI: 10.1186/s13045-025-01698-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Gastric cancer is the fifth most common cancer globally and is associated with significant morbidity and mortality. Despite its alarming prevalence, limited comparative evidence exists on its treatment efficacy and prognosis across diverse China populations. METHODS To address this, our study used a large-scale dataset from the National Cancer Information Database, including data from 220,304 patients from 53 leading hospitals across 27 provinces in China. RESULTS From 2017 to 2023, early-stage (Stages I-II) gastric cancer diagnoses increased to 35.63% of all cancer cases. Our study evaluated the neoadjuvant treatment strategies, adjuvant post-operative therapy, first- and second-line management for progressive stages, alongside current gastric cancer treatment guidelines in China. Notably, immunotherapy accounted for 16.17% and 23.28% of first- and second-line treatments for late-stage gastric cancers, and 14.56% and 5.00% for neoadjuvant and adjuvant therapies, respectively. Analysis of survival rates revealed that the 1-, 2-, 3-, 4-, and 5-year survival rates were 74.07%, 54.89%, 44.21%, 37.97%, and 33.53%, respectively. The 5-year survival rates across stages I-IV were 85.07%, 49.34%, 35.56%, and 13.15%, respectively. CONCLUSIONS These findings offer critical insights into the current state of gastric cancer treatment in China and can inform future initiatives to improve therapeutic outcomes for patients with gastric cancer.
Collapse
Affiliation(s)
- Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Keren Jia
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yi Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jiajia Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Dan Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Lei Jiang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Haoxin Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | | | - Jian Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
246
|
Duan G, Qi M, Xun L, An Y, Zuo Z, Luo Y, Song Z. Metformin Enhances the Chemosensitivity of Gastric Cancer to Cisplatin by Downregulating Nrf2 Level. Anal Cell Pathol (Amst) 2025; 2025:5714423. [PMID: 40264514 PMCID: PMC12014253 DOI: 10.1155/ancp/5714423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 04/24/2025] Open
Abstract
Cisplatin-based chemotherapy resistance is a common issue for cancer clinical efficacy. Metformin is being studied for its possible anticancer effect. The present study aimed to investigate whether metformin affects the chemosensitivity of gastric cancer to cisplatin and reveal the molecular mechanism. In this study, the effects of combination therapy with metformin and cisplatin on cell viability, cell apoptosis, malondialdehyde, superoxide dismutase, reactive oxygen species level, glucose uptake, lactate production, protein level, and xenograft tumor formation were analyzed in gastric cancer cells. Immunohistochemical staining was performed to detect Ki67 expression in matched tumor samples. The results showed that NCI-N87 and SNU-16 cells were most resistant and sensitive to cisplatin, respectively. Metformin treatment increased the cisplatin sensitivity of gastric cancer by inhibiting cell viability and metabolic reprogramming and promoting cell apoptosis and oxidative stress. Furthermore, overexpression of nuclear factor erythroid 2-related factor 2 (Nrf2) reversed the effects of metformin in the cisplatin sensitivity of gastric cancer by inhibiting cell viability and metabolic reprogramming and promoting cell apoptosis and oxidative stress. Metformin activated p53 and AMPK pathways in cisplatin-induced NCI-N87 cells, which were reversed by upregulating Nrf2. BAY-3827 (AMPK inhibitor) or p-nitro-Pifithrin-α (p53 inhibitor) treatments also reversed the effects of metformin increased the cisplatin sensitivity of gastric cancer by inhibiting cell viability and metabolic reprogramming and promoting cell apoptosis and oxidative stress. These results suggest that metformin significantly increases chemosensitivity of gastric cancer to cisplatin by inhibiting Nrf2 expression and metabolic reprogramming and activating oxidative stress and the pathway of p53 and AMPK.
Collapse
Affiliation(s)
- Guihua Duan
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Min Qi
- Department of Radiology, The Third People's Hospital of Kunming City, The Sixth Affiliated Hospital of Dali University, Kunming, Yunnan, China
| | - Linting Xun
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Ying An
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Zan Zuo
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yusi Luo
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Zhengji Song
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
247
|
Zhang F, Wang B, Wu M, Zhang L, Ji M. Current status of KRAS G12C inhibitors in NSCLC and the potential for combination with anti-PD-(L)1 therapy: a systematic review. Front Immunol 2025; 16:1509173. [PMID: 40303413 PMCID: PMC12037499 DOI: 10.3389/fimmu.2025.1509173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
In recent years, precision medicine for non-small cell lung cancer (NSCLC) has made significant strides, particularly with advancements in diagnostic and therapeutic technologies. Targeted 7therapies and Anti-PD-(L)1 Therapies have emerged as vital treatment options, yet KRAS mutations, especially KRAS G12C, have been historically difficult to address. Due to the unique activation mechanism of KRAS G12C has led to the development of specific inhibitors, such as AMG 510 and MRTX849, which show promising therapeutic potential. However, results from the CodeBreaK 200 Phase III trial indicated that AMG 510 did not significantly improve overall survival compared to docetaxel. Resistance after prolonged use of KRAS G12C inhibitors continues to pose a challenge, prompting interest in new drugs and combination strategies. KRAS mutations can impair tumor-infiltrating T cell function and create an immunosuppressive tumor microenvironment, making the combination of KRAS G12C inhibitors with anti-PD-(L)1 therapies particularly appealing. Preliminary data suggest these combinations may enhance both survival and quality of life, though safety concerns remain a barrier. Ongoing research is crucial to refine treatment regimens and identify suitable patient populations. This review focuses on the development of KRAS G12C inhibitors in monotherapy and combination therapies for NSCLC, discussing major clinical trials and future research directions.
Collapse
Affiliation(s)
| | | | | | | | - Mei Ji
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
248
|
Lamprecht CB, Kashuv T, Lucke-Wold B. Understanding metastatic patterns in gastric cancer: Insights from lymph node distribution and pathology. World J Gastrointest Oncol 2025; 17:103709. [PMID: 40235890 PMCID: PMC11995340 DOI: 10.4251/wjgo.v17.i4.103709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/26/2025] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Gastric cancer (GC) represents a significant global health burden due to its high morbidity and mortality. Specific behaviors of GC sub-types, distinct dissemination patterns, and associated risk-factors remain poorly understood. This editorial highlights several key prognostic factors, including pathological staging and vascular invasion, that impact GC. It examines a recent study's investigation of differential metastatic lymph nodes distribution and survival in upper and lower GC sub-types, focusing on histological characterization, pathophysiology, usage of neoadjuvant chemotherapy, and additional predictive determinants. We assess the statistical robustness and clinical applicability of the findings, underscoring the importance of treating GC as a heterogeneous disease and emphasizing how tailored surgical approaches informed by lymph node distribution can optimize tumor detection while minimizing unnecessary interventions. The study's large cohort, multi-center design, and strict inclusion criteria strengthen its validity in guiding surgical planning and risk-stratification. However, integrating genetic and molecular data is critical for refining models and broadening applicability. Additionally, recurrence-metrics and infection-related factors, such as Helicobacter pylori and Epstein-Barr virus, absent in the original study, remain vital for directing future research. By bridging metastatic patterns with prospective methodologies and inclusion of diverse populations, this editorial provides a framework for advancing early detection and personalized GC care.
Collapse
Affiliation(s)
- Chris B Lamprecht
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL 32608, United States
| | - Tyler Kashuv
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL 32608, United States
| | - Brandon Lucke-Wold
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL 32608, United States
| |
Collapse
|
249
|
Evrard C, Rochelle T, Martel M, Al Achkar A, Ferru A, Randrian V, Karayan-Tapon L, Tougeron D. Prognostic and predictive value of microsatellite instability analysis in ctDNA by digital droplet PCR for patients with MSI colorectal cancers. J Transl Med 2025:104176. [PMID: 40246285 DOI: 10.1016/j.labinv.2025.104176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Deficient MisMatch Repair (dMMR) and/or microsatellite instability (MSI) colorectal cancer (CRC) is highly sensitive to immune checkpoint inhibitors (ICI). It is thus becoming increasingly relevant to monitor circulating tumor DNA (ctDNA) and to determine MSI status (ctDNA-MSI) in CRC. So far, few studies have explored this, even though it could be particularly relevant in evaluating treatment efficacy in dMMR and/or MSI CRC patients. The ctDNA digestive cancers study (ADI-MSI) aims to assess the value of ctDNA-MSI as a predictor of ICI efficacy. Blood samples were collected prospectively in a single-center cohort to analyse circulating cell-free DNA (cfDNA) and ctDNA-MSI before the start and during treatment. ctDNA-MSI was measured using digital droplet PCR with the five microsatellite markers of the Pentaplex panel. The primary endpoint was to evaluate ctDNA-MSI levels as a predictor of progression-free survival (PFS). We included 54 dMMR and/or MSI CRC patients, most of whom had metastatic disease (77.8%) treated in the first (25.9%) or second-line (42.6%) with ICI. High baseline cfDNA and ctDNA-MSI were associated with worse PFS and worse overall survival (OS). ctDNA-MSI kinetics, but not cfDNA kinetics, was associated with treatment response (p=0.006), PFS (p=0.03) and OS (p=0.04). ctDNA-MSI kinetics divided into three groups (increase, decrease and negative) correlated strongly with PFS (PFS at 24 months was 0%, 53.0% and 77.0%, respectively; p<0.001) and remained significant in multivariate analysis (HR=7.93; 95% CI, 2.23-28.21; p=0.005). Since there is no strong predictor of ICI efficacy in dMMR and/or MSI CRC patients, these results suggest that ctDNA-MSI could help physicians in treatment decision-making in the future.
Collapse
Affiliation(s)
- Camille Evrard
- Université de Poitiers, PRoDiCeT, Poitiers, France; CHU de Poitiers, Service d'Oncologie Médicale, Poitiers, France
| | - Tristan Rochelle
- CHU de Poitiers, Service de Cancérologie Biologique, Poitiers, France
| | - Marine Martel
- CHU de Poitiers, Service de Cancérologie Biologique, Poitiers, France
| | - Anis Al Achkar
- Université de Poitiers, PRoDiCeT, Poitiers, France; CHU de Poitiers, Service de Cancérologie Biologique, Poitiers, France
| | - Aurélie Ferru
- CHU de Poitiers, Service d'Oncologie Médicale, Poitiers, France
| | - Violaine Randrian
- Université de Poitiers, PRoDiCeT, Poitiers, France; CHU de Poitiers, Service d'Hépato-Gastro-Entérologie, Poitiers, France
| | - Lucie Karayan-Tapon
- Université de Poitiers, PRoDiCeT, Poitiers, France; CHU de Poitiers, Service de Cancérologie Biologique, Poitiers, France
| | - David Tougeron
- Université de Poitiers, PRoDiCeT, Poitiers, France; CHU de Poitiers, Service d'Hépato-Gastro-Entérologie, Poitiers, France.
| |
Collapse
|
250
|
An S, Liu FT, Wang C. Comment on "Phase 2 study of neoadjuvant durvalumab plus docetaxel, oxaliplatin, and S-1 with surgery and adjuvant durvalumab plus S-1 for resectable locally advanced gastric cancer". J Immunother Cancer 2025; 13:e012221. [PMID: 40234088 PMCID: PMC12001348 DOI: 10.1136/jitc-2025-012221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/17/2025] Open
Affiliation(s)
- Shuai An
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Fang-Tong Liu
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| | - Chenglong Wang
- Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
| |
Collapse
|